Slot System
Featured Buckets
Featured Buckets Admin

Family and Caregiver Needs Over the Course of the Cancer Trajectory

Article Type
Changed
Fri, 01/04/2019 - 11:04

When a patient is diagnosed with cancer, family members often assume responsibility for providing care. They are typically involved not only with the diagnostic and treatment phases of care but also across the care trajectory and into survivorship. These caregivers are a primary source of support to individuals with cancer. The purpose of this article is to present an overview of the challenges, needs, and roles of family caregivers over the course of the cancer treatment trajectory and to discuss what support the professionals can provide.

Click of the PDF icon at the top of this introduction to read the full article.

Author and Disclosure Information

 

 

Publications
Topics
Sections
Author and Disclosure Information

 

 

Author and Disclosure Information

 

 

When a patient is diagnosed with cancer, family members often assume responsibility for providing care. They are typically involved not only with the diagnostic and treatment phases of care but also across the care trajectory and into survivorship. These caregivers are a primary source of support to individuals with cancer. The purpose of this article is to present an overview of the challenges, needs, and roles of family caregivers over the course of the cancer treatment trajectory and to discuss what support the professionals can provide.

Click of the PDF icon at the top of this introduction to read the full article.

When a patient is diagnosed with cancer, family members often assume responsibility for providing care. They are typically involved not only with the diagnostic and treatment phases of care but also across the care trajectory and into survivorship. These caregivers are a primary source of support to individuals with cancer. The purpose of this article is to present an overview of the challenges, needs, and roles of family caregivers over the course of the cancer treatment trajectory and to discuss what support the professionals can provide.

Click of the PDF icon at the top of this introduction to read the full article.

Publications
Publications
Topics
Article Type
Sections
PURLs Copyright

Disallow All Ads
Alternative CME
Use ProPublica

Coordination of Care in Breast Cancer Survivors: An Overview

Article Type
Changed
Thu, 12/15/2022 - 18:26
Display Headline
Coordination of Care in Breast Cancer Survivors: An Overview

 

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215

doi:10.1016/j.suponc.2011.06.008 | How to Cite or Link Using DOI
  Permissions & Reprints

How We Do It

Coordination of Care in Breast Cancer Survivors: An Overview

Kimberly S. Peairs MD 
, Antonio C. Wolff MD, FACP, Sharon J. Olsen PhD, Elissa T. Bantug MHS, Lillie Shockney RN, BS, MAS, Melinda E. Kantsiper MD, Elisabeth Carrino-Tamasi MSW, LGSW, Claire F. Snyder PhD

[Author vitae]

Received 3 March 2011; Accepted 18 June 2011. Available online 3 November 2011.

TO READ THE ENTIRE ARTICLE, CLICK ON THE ADJACENT LINK TO THE PDF FILE

Abstract

The number of breast cancer survivors in the United States is increasing. With longer survival, there has been an increase in the complexity and duration of posttreatment care. Multidisciplinary care teams are needed to participate across the broad spectrum of issues that breast cancer survivors face. In this setting, the need for well-established patterns of communication between care providers is increasingly apparent. We have created a multidisciplinary approach to the management of breast cancer survivors to improve communication and education between providers and patients. This approach could be extended to the care and management of survivors of other types of cancer.

Case

A 65-year-old woman with stage II breast cancer, mild hypertension, and obesity recently completed treatment for her estrogen/progesterone receptor–positive, HER 2–negative breast cancer. She was treated with lumpectomy, radiation therapy, and adjuvant chemotherapy with doxorubicin and cyclophosphamide followed by paclitaxel. She remains on an aromatase inhibitor and is experiencing arthralgias, numbness in her extremities, fatigue, and apprehension about cancer recurrence. She has not seen her primary care physician since the start of her cancer treatment but is concerned that her “heart” and bones may be affected by her therapy.

Vitae

Dr. Peairs is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Wolff is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Olsenis from the Johns Hopkins School of Nursing, Baltimore, Maryland.

Dr. Bantugis from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Shockney is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Kantsiper is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Carrino-Tamasi is from the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Snyder is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215
Article PDF
Author and Disclosure Information

Publications
Topics
Legacy Keywords
breast cancer survivors
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

 

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215

doi:10.1016/j.suponc.2011.06.008 | How to Cite or Link Using DOI
  Permissions & Reprints

How We Do It

Coordination of Care in Breast Cancer Survivors: An Overview

Kimberly S. Peairs MD 
, Antonio C. Wolff MD, FACP, Sharon J. Olsen PhD, Elissa T. Bantug MHS, Lillie Shockney RN, BS, MAS, Melinda E. Kantsiper MD, Elisabeth Carrino-Tamasi MSW, LGSW, Claire F. Snyder PhD

[Author vitae]

Received 3 March 2011; Accepted 18 June 2011. Available online 3 November 2011.

TO READ THE ENTIRE ARTICLE, CLICK ON THE ADJACENT LINK TO THE PDF FILE

Abstract

The number of breast cancer survivors in the United States is increasing. With longer survival, there has been an increase in the complexity and duration of posttreatment care. Multidisciplinary care teams are needed to participate across the broad spectrum of issues that breast cancer survivors face. In this setting, the need for well-established patterns of communication between care providers is increasingly apparent. We have created a multidisciplinary approach to the management of breast cancer survivors to improve communication and education between providers and patients. This approach could be extended to the care and management of survivors of other types of cancer.

Case

A 65-year-old woman with stage II breast cancer, mild hypertension, and obesity recently completed treatment for her estrogen/progesterone receptor–positive, HER 2–negative breast cancer. She was treated with lumpectomy, radiation therapy, and adjuvant chemotherapy with doxorubicin and cyclophosphamide followed by paclitaxel. She remains on an aromatase inhibitor and is experiencing arthralgias, numbness in her extremities, fatigue, and apprehension about cancer recurrence. She has not seen her primary care physician since the start of her cancer treatment but is concerned that her “heart” and bones may be affected by her therapy.

Vitae

Dr. Peairs is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Wolff is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Olsenis from the Johns Hopkins School of Nursing, Baltimore, Maryland.

Dr. Bantugis from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Shockney is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Kantsiper is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Carrino-Tamasi is from the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Snyder is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215

 

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215

doi:10.1016/j.suponc.2011.06.008 | How to Cite or Link Using DOI
  Permissions & Reprints

How We Do It

Coordination of Care in Breast Cancer Survivors: An Overview

Kimberly S. Peairs MD 
, Antonio C. Wolff MD, FACP, Sharon J. Olsen PhD, Elissa T. Bantug MHS, Lillie Shockney RN, BS, MAS, Melinda E. Kantsiper MD, Elisabeth Carrino-Tamasi MSW, LGSW, Claire F. Snyder PhD

[Author vitae]

Received 3 March 2011; Accepted 18 June 2011. Available online 3 November 2011.

TO READ THE ENTIRE ARTICLE, CLICK ON THE ADJACENT LINK TO THE PDF FILE

Abstract

The number of breast cancer survivors in the United States is increasing. With longer survival, there has been an increase in the complexity and duration of posttreatment care. Multidisciplinary care teams are needed to participate across the broad spectrum of issues that breast cancer survivors face. In this setting, the need for well-established patterns of communication between care providers is increasingly apparent. We have created a multidisciplinary approach to the management of breast cancer survivors to improve communication and education between providers and patients. This approach could be extended to the care and management of survivors of other types of cancer.

Case

A 65-year-old woman with stage II breast cancer, mild hypertension, and obesity recently completed treatment for her estrogen/progesterone receptor–positive, HER 2–negative breast cancer. She was treated with lumpectomy, radiation therapy, and adjuvant chemotherapy with doxorubicin and cyclophosphamide followed by paclitaxel. She remains on an aromatase inhibitor and is experiencing arthralgias, numbness in her extremities, fatigue, and apprehension about cancer recurrence. She has not seen her primary care physician since the start of her cancer treatment but is concerned that her “heart” and bones may be affected by her therapy.

Vitae

Dr. Peairs is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Wolff is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Olsenis from the Johns Hopkins School of Nursing, Baltimore, Maryland.

Dr. Bantugis from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Shockney is from the Johns Hopkins School of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Kantsiper is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

Dr. Carrino-Tamasi is from the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.

Dr. Snyder is from the Johns Hopkins School of Medicine, Baltimore, Maryland.

The Journal of Supportive Oncology
Volume 9, Issue 6, November-December 2011, Pages 210-215
Publications
Publications
Topics
Article Type
Display Headline
Coordination of Care in Breast Cancer Survivors: An Overview
Display Headline
Coordination of Care in Breast Cancer Survivors: An Overview
Legacy Keywords
breast cancer survivors
Legacy Keywords
breast cancer survivors
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Guiding Patients Facing Decisions about “Futile” Chemotherapy

Article Type
Changed
Fri, 01/04/2019 - 11:04
Display Headline
Guiding Patients Facing Decisions about “Futile” Chemotherapy

The Journal of Supportive Oncology
Volume 9, Issue 5, September-October 2011, Pages 184-187


doi:10.1016/j.suponc.2011.04.001 
  Permissions & Reprints

How We Do It

Guiding Patients Facing Decisions about “Futile” Chemotherapy

Erin Alesi MD, Barton Bobb RN, MSN, Thomas J. Smith MD 

Received 4 April 2011; Accepted 16 December 2011. Available online 24 September 2011.

Case Presentation

Ms. G is a 71-year-old woman with metastatic gastric adenocarcinoma recently diagnosed after an extensive surgical resection for a small bowel obstruction (SBO). She was admitted from the surgery clinic with intractable nausea and vomiting. An abdominal computerized tomographic (CT) scan revealed a partial SBO and peritoneal carcinomatosis. Given her recent surgery, the extent of her disease, and high likelihood of recurrent SBO, the surgical team decided that Ms. G was no longer a surgical candidate. When her symptoms did not improve with conservative measures, both oncology and palliative medicine were consulted to assist with symptom management and goals of care. The oncology team stated that Ms. G was still a chemotherapy candidate and suggested that she attend her new patient evaluation in oncology clinic the following week. The palliative medicine team then met with the patient to discuss management options and her preferences for care. The palliative care team explained ways to control her nausea and vomiting without using a nasogastric tube, and the patient agreed to transfer to their service for symptom management. The palliative team explained that her cancer was incurable but that chemotherapy options existed to help control her disease and possibly prolong her life. They also explained that the chemotherapy has side effects and that the patient would need to decide if she wanted to undergo treatment and accept potential side effects for the possibility of prolonging her life by weeks to months and improving her symptoms. As an alternative, she was told that she could focus solely on symptom control with medications and allow her disease to take its natural course. Ms. G was asked to think about how she wanted to spend the time she had left. Prior to discharge, as her symptoms improved, Ms. G was evaluated by another oncologist, who, after consulting the expert gastrointestinal cancer team, explained to her that the current chemotherapy options available for metastatic gastric cancer were rarely, if ever, successful at reversing malignant obstruction. With this information, the patient decided to be discharged home with hospice and spend time with her family. She died peacefully at her home approximately two weeks later.

Article Outline

Futile Is as Futile Does

When deciding whether or not chemotherapy is “futile,” the concept of medical futility must be explored.[1] Though it remains difficult to adequately define, the qualitative and quantitative descriptions offered by Schneiderman et al[2] are widely used. Qualitatively, futile treatment “merely preserves permanent unconsciousness or cannot end dependence on intensive medical care.” More precisely, it is a medical treatment “that in the last 100 cases … has been useless.”[2] A useful, albeit imprecise, definition of futile chemotherapy is that in which the burdens and risks outweigh the benefits. As an example, studies on chemotherapy for advanced non-small-cell lung cancer (NSCLC) have shown that patients with poor performance status or chemotherapy-unresponsive disease receive little benefit in terms of response rates and survival. [3] and [4] A retrospective analysis by Massarelli et al3 showed dismal response rates for third- and fourth-line NSCLC chemotherapy of 2.3% and 0%, respectively. Additionally, an observational study by Zietemann and Duell[4] showed that 40% and 50% of patients receiving second- and third-line chemotherapy for NSCLC die during or soon after treatment, respectively, and that over 20% receive chemotherapy within 14 days of death. Neither study commented on quality of life experienced by patients. However, a recent study by Temel et al[5] demonstrated that NSCLC patients receiving concurrent palliative care and standard oncologic care had better quality of life and even longer survival than patients receiving only standard oncologic care, despite being less likely to receive aggressive end-of-life care. Though limited to patients with NSCLC, these studies illustrate that chemotherapy in advanced cancer is often futile, especially when less aggressive care can improve quality of life as well as survival.

 

 

Addressing the futility of chemotherapy with patients is challenging for most oncologists. Although defining treatments as “futile” is suitable in the medical literature, it is a word that may carry negative connotations, such as hopelessness or abandonment, to patients. A more descriptive and less negative term, “nonbeneficial,” may be used when discussing futile chemotherapy with patients. The point when chemotherapy becomes nonbeneficial, and thus futile, is different for each patient and might even change over time. Addressing the patient's definition of nonbeneficial chemotherapy regularly during treatment ensures that the patient's goals are clear and allows the oncologist to direct conversation toward alternative options, such as palliative and hospice care, when chemotherapy cannot provide the benefits sought by the patient. This can be as simple as asking the patient, “Do you think the chemotherapy is giving you enough benefit to continue?”

Palliative Care: It's Not Just Giving Up on People

Both the physician and the patient face several decisions when considering whether or not to pursue chemotherapy for advanced cancer. First of all, the patient must decide how much information he or she wants from the oncologist. If the patient is the decision maker, he or she must choose to accept chemotherapy that is palliative, not curative. After a frank discussion about the anticipated outcomes and symptoms associated with chemotherapy, the patient must consider whether he or she can accept the burden of treatment for the potential of prolonging life by days, weeks, or months. On the other hand, the oncologist must decide if chemotherapy should even be offered, based on patient performance status, known therapeutic outcomes, and patient values and goals. The oncologist can reassure patients that the best available data show that patients who use hospice for even one day actually live longer than those who do not.[6] Once informed about what palliative care and hospice offer, the patient may determine whether or not alternatives to chemotherapy are more favorable. If the patient qualifies for clinical trials, he or she must decide to accept treatment with uncertain outcome. When reflecting upon such difficult issues, both the patient and oncologist should involve others to help guide decision making. Oncologists can consult trusted colleagues for their expertise and to ensure that they are using the best information available. Patients should involve loved ones whom they trust to help make decisions in their best interest. Table 1 provides key questions that the oncologist faces when making these decisions and how to approach them.

Table 1: Questions to discuss with the patient when chemotherapy may be futile

Question

Leading prompts

Comment

What is the patient’s current understanding of the disease?

How much do you know about your cancer at this point?

 

 

How much do you want to know?

Be sure the patient is ready to discuss this issue and that you have enough time for discussion.

 

Ask if there are others who should receive this information simultaneously, afterwards, or instead of the patient.

What are the patient’s goals?

Knowing that we can’t cure your cancer, what are your goals, wishes, or hopes for the future?

Treatment decisions may be impacted greatly by a patient’s personal goals (e.g. patient wants to live to child’s graduation, or patient wants to be as comfortable as possible)

If chemotherapy is an option and the patient is interested, is he/she aware of potential risks and benefits?

Although everyone responds differently, these are the likely side effects and outcomes of this treatment…

Be specific in terms of likelihood of response, type of response (palliation instead of cure, extent of life prolongation expected, symptom relief, etc.) and how likely it is that treatment will help achieve patient’s goals.

 

Discuss potential symptom burden from treatment in detail.

 

Patient needs to be able to make informed decision about risks vs. benefits involved in potential treatment.

If the patient declines chemotherapy, treatment is not indicated, or treatment fails, what other options are available?

Let’s talk about options to make sure that you are comfortable and enjoy the highest quality of life possible in the time that you have left.

Focus on pain and symptom management. Discuss hospice options (home vs. inpatient) and make referrals when appropriate.

 

Stress that you will continue your relationship with the patient (possibly as their hospice provider) and that you will ensure that their symptoms are managed, either directly or through hospice nurses.

 

 

As an alternative to addressing the above issues with the patient independently, oncologists may involve a palliative care specialist to facilitate this conversation.[7] Particularly in cases where the oncologist decides that chemotherapy is no longer a viable option, it may be easier, from both the patient and the provider perspectives, for the palliative care specialist to have this discussion. In a recent survey of patients on our oncology ward, the great majority did not want to discuss advance directives (ADs) with their oncologist—these patients thought ADs were important and should be discussed but were more comfortable discussing them with the admitting provider than the oncologist.[8] Patients may feel that they are disappointing their oncologist by being unable to take further treatment or by admitting that treatment has failed them. Similarly, oncologists might view having this discussion as an admission of their failure as a provider. The palliative care specialist, on the other hand, has no responsibility for chemotherapy and possibly no prior relationship with the patient, thus alleviating this type of emotional association between provider and patient. Furthermore, the conversation about nonbeneficial chemotherapy provides a segue for the palliative care provider to discuss with patients what he or she does best: establishing goals of care, managing symptoms, and maintaining comfort. For the palliative care specialist, providing symptom management and the best possible quality of life for patients are the fundamental goals. Death is generally not viewed with a sense of failure when palliation is the focus of care.

Oncology: Palliative Care Is Giving Up

We still hear from oncologists like ourselves the dreaded words “What do you want me to do, give up on the patient?” or, to the patient, “What, are you giving up? I thought you'd keep fighting!” We would argue that current best practices include knowing when the risks and harms of chemotherapy outweigh any potential chance of benefit. Physicians and patients should follow current National Comprehensive Cancer Network (NCCN) guidelines for solid tumors such as breast9 and lung10 cancer and stop chemotherapy when the chance of success is minimal. If the doctor cannot describe a specific, substantial benefit that outweighs the toxicity, he or she should not recommend it.[11] And all the relevant guidelines call for considering a switch to nonchemotherapy palliative care when the patient's performance status is Eastern Cooperative Oncology Group (ECOG) ≥3, defined as “3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours.”[12] Such a simple threshold could dramatically reduce the use of chemotherapy at the end of life and lessen downstream toxicities.

Oncologists can implement several strategies to help facilitate the transition from aggressive care to comfort care (Table 2). For patients with incurable cancer, oncologists can hold early discussions about palliative and hospice options that will need to be implemented when chemotherapy is no longer able to control their disease. This discussion introduces palliative medicine as part of the care plan for incurable disease and allows the patient to anticipate such a transition. Oncologists can also provide reassurance that they will continue to be involved in their patient's care and to support them, even if the patient does not undergo further chemotherapy. There are at least four studies that show equal[13] or better[6] survival, smoother transitions to hospice when death is inevitable, less intensive end-of-life care, and superior patient and family outcomes with concurrent palliative care. [14] and [15] By helping patients establish legal documents, such as ADs and power of attorney, oncologists and palliative care specialists can alleviate some of the stress related to the end of life and make the transition to comfort care easier. Finally, oncologists can review guidelines such as those from the NCCN and American Society of Clinical Oncology, which call for a switch to palliative care when the cancer has grown on three regimens or the patient's ECOG performance status is three or above. [11] and [12]

Table 2: Things that help ONCOLOGISTS and their patients

Item

How it helps

Comments

Early discussion of palliative and hospice care when chemotherapy may no longer help.

Hospice (and eventual death) will not come as a complete surprise.

“We will do our best to help you with this cancer, but at some point there may not be any treatments known to help….” “Remember the conversation we had when we first met?...”

Reassurance that the oncologist will not abandon the patient if concurrent care is given.

This major fear may keep oncology patients at the same practice they have known for years – it is familiar – when they would be better served by transition.

There are now at least 4 randomized trials showing that most patients will accept concurrent palliative care if offered, and that outcomes are equal or better, at less cost.6,13,14,15

Legal documents such as Advance Medical Directives, Durable Medical Power of Attorney

Reinforces the seriousness and “now” aspect of care.

These are readily available in all states at no cost. They are not the final word on how to live one’s remaining time, but will get the conversation started.

Best nationally recognized information showing that further chemotherapy will not help due to 3 prior failures, or is not indicated due to poor performance status.9,10

The oncologist can point to the right page and say “The best national guidelines call for a switch away from chemo…because it will do no good and will cause harmful side effects.”

Readily accessed from the Internet.

Use decision aids, similar to Adjuvant!.

Increases the amount of truthful information given, even when the news is bad, and helps with transition points.

An increasing number of these are available[i],[ii],[iii],[iv] and will soon be offered as smart phone applications (aps).

 

 

Communication tools, such as the National Cancer Institute's Oncotalk and EPEC-O, are useful for oncologists seeking to further enhance their communication skills.

Take-Home Messages

Guiding patients in making decisions about nonbeneficial, or futile, chemotherapy presents a challenge for many oncologists as well as their patients and families. Though futility is difficult to define, oncologists and their patients can decide through regular, open discussion if the burdens of chemotherapy outweigh the benefits and whether or not chemotherapy can achieve the reasonable benefits desired by the patient. “Your cancer is advancing despite our best efforts to keep it from growing. Let's talk about what options we have at this point and see what will work best for you.” To make such decisions, oncologists must obtain the most current information and convey it to patients (or their designated decision makers) as clearly as possible. “Based on the latest evidence, there is a 20% chance that the cancer will shrink or stay the same size with this treatment and an 80% chance that it will continue to grow despite treatment.” Both oncologists and their patients should involve those whom they trust to help with decision making. In cases where chemotherapy is nonbeneficial, oncologists may prefer to involve palliative and hospice care specialists to discuss the transition to comfort care with the patient. “At this time, I do not have any treatments that are likely to help you live longer or more comfortably, but I want to make sure that we get the most out of the rest of your life. I have asked a palliative care specialist to help us make this possible.” In order to ease the transition from aggressive or curative care to comfort care, oncologists can employ approaches such as early discussion of palliative and hospice care, assuring the patient of continued involvement in their care, and helping patients with ADs. These approaches not only benefit patients and their families but also strengthen the relationship between the oncologist and the patients and their families.

Acknowledgments

This research was supported by grants GO8 LM0095259 from the National Library of Medicine and R01CA116227-01 (both to T. J. S.) from the National Cancer Institute.

References [PubMed ID in brackets]

1 P.R. Helft, M. Siegler and J. Lantos, The rise and fall of the medical futility movement, . N Engl J Med,  343  (2000), pp. 293–296 [10911014].

2 L.J. Schneiderman, N.S. Jecker and A.R. Jonsen, Medical futility: its meaning and ethical implications, . Ann Intern Med,  112  (1990), pp. 949–954 [2187394].

3 E. Massarelli, F. Andre, D.D. Liu, J.J. Lee, M. Wolf, A. Fandi, J. Ochs, T. Le Chevalier, F. Fossella and R.S. Herbst, A retrospective analysis of the outcome of patients who have received two prior chemotherapy regimens including platinum and docetaxel for recurrent non-small-cell lung cancer, . Lung Cancer,  39 1 (2003), pp. 55–61 [12499095].

4 V. Zietemann and T. Duell, Every-day clinical practice in patients with advanced non-small-cell lung cancer, . Lung Cancer,  68 2 (2010), pp. 273–277 [19632737].

5 J.S. Temel, J.A. Greer, A. Muzikansky, E.R. Gallagher, S. Admane, V.A. Jackson, C.M. Dahlin, C.D. Blinderman, J. Jacobsen, W.F. Pirl, J.A. Billings and T.J. Lynch, Early palliative care for patients with metastatic non-small-cell lung cancer, . N Engl J Med,  363  (2010), pp. 733–742 [20818875].

6 S.R. Connor, B. Pyenson, K. Fitch, C. Spence and K. Iwasaki, Comparing hospice and nonhospice patient survival among patients who die within a three-year window. J Pain Symptom Manage,  33 3 (2007), pp. 238–246.

7 S.E. Harrington and T.J. Smith, The role of chemotherapy at the end of life: “when is enough, enough?”. JAMA,  299 22 (2008), pp. 2667–2678.

8 L. Dow, R. Matsuyama, L. Kuhn, L. Lyckholm, E.B. Lamont and T.J. Smith, Paradoxes in advance care planning. J Clin Oncol,  28 2 (2010), pp. 299–304.

9 NCCN Breast Cancer Clinical Practice Guidelines Panel, R.W. Carlson, D.C. Allred, B.O. Anderson, H.J. Burstein, W.B. Carter, S.B. Edge, J.K. Erban, W.B. Farrar, L.J. Goldstein, W.J. Gradishar, D.F. Hayes, C.A. Hudis, M. Jahanzeb, K. Kiel, B.M. Ljung, P.K. Marcom, I.A. Mayer, B. McCormick, L.M. Nabell, L.J. Pierce, E.C. Reed, M.L. Smith, G. Somlo, R.L. Theriault, N.S. Topham, J.H. Ward, E.P. Winer and A.C. Wolff, Breast cancer. J Natl Compr Canc Netw,  7 2 (2009), pp. 122–192.

10 NCCN Non-Small Cell Lung Cancer Panel Members, D.S. Ettinger, W. Akerley, G. Bepler, M.G. Blum, A. Chang, R.T. Cheney, L.R. Chirieac, T.A. D'Amico, T.L. Demmy, A.K. Ganti, R. Govindan, F.W. Grannis, T. Jahan, M. Jahanzeb, D.H. Johnson, A. Kessinger, R. Komaki, F.M. Kong, M.G. Kris, L.M. Krug, Q.T. Le, I.T. Lennes, R. Martins, J. O'Malley, R.U. Osarogiagbon, G.A. Otterson, J.D. Patel, K.M. Pisters, K. Reckamp, G.J. Riely, E. Rohren, G.R. Simon, S.J. Swanson, D.E. Wood and S.C. Yang, Non-small cell lung cancer. J Natl Compr Canc Netw,  8 7 (2010), pp. 740–801.

 

 

11 American Society of Clinical Oncology Outcomes Working Group, Outcomes of cancer treatment for technology assessment and cancer treatment guidelines. J Clin Oncol,  14  (1996), pp. 671–679.

12 Eastern Cooperative Oncology Group, ECOG Performance Status, http://ecog.dfci.harvard.edu/general/perf_stat.html Accessed November 30, 2010. 

13 J. Finn, K. Pienta and J. Parzuchowski, Bridging cancer treatment and hospice care. Proc Am Soc Clin Oncol,  21  (2002), p. 1452. 

14 G. Gade, I. Venohr, D. Conner, K. McGrady, J. Beane, R.H. Richardson, M.P. Williams, M. Liberson, M. Blum and R. Della Penna, Impact of an inpatient palliative care team: a randomized control trial. J Palliat Med,  11 2 (2008), pp. 180–190.

Copyright © 2011 Elsevier Inc. All rights reserved.


Article PDF
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

The Journal of Supportive Oncology
Volume 9, Issue 5, September-October 2011, Pages 184-187


doi:10.1016/j.suponc.2011.04.001 
  Permissions & Reprints

How We Do It

Guiding Patients Facing Decisions about “Futile” Chemotherapy

Erin Alesi MD, Barton Bobb RN, MSN, Thomas J. Smith MD 

Received 4 April 2011; Accepted 16 December 2011. Available online 24 September 2011.

Case Presentation

Ms. G is a 71-year-old woman with metastatic gastric adenocarcinoma recently diagnosed after an extensive surgical resection for a small bowel obstruction (SBO). She was admitted from the surgery clinic with intractable nausea and vomiting. An abdominal computerized tomographic (CT) scan revealed a partial SBO and peritoneal carcinomatosis. Given her recent surgery, the extent of her disease, and high likelihood of recurrent SBO, the surgical team decided that Ms. G was no longer a surgical candidate. When her symptoms did not improve with conservative measures, both oncology and palliative medicine were consulted to assist with symptom management and goals of care. The oncology team stated that Ms. G was still a chemotherapy candidate and suggested that she attend her new patient evaluation in oncology clinic the following week. The palliative medicine team then met with the patient to discuss management options and her preferences for care. The palliative care team explained ways to control her nausea and vomiting without using a nasogastric tube, and the patient agreed to transfer to their service for symptom management. The palliative team explained that her cancer was incurable but that chemotherapy options existed to help control her disease and possibly prolong her life. They also explained that the chemotherapy has side effects and that the patient would need to decide if she wanted to undergo treatment and accept potential side effects for the possibility of prolonging her life by weeks to months and improving her symptoms. As an alternative, she was told that she could focus solely on symptom control with medications and allow her disease to take its natural course. Ms. G was asked to think about how she wanted to spend the time she had left. Prior to discharge, as her symptoms improved, Ms. G was evaluated by another oncologist, who, after consulting the expert gastrointestinal cancer team, explained to her that the current chemotherapy options available for metastatic gastric cancer were rarely, if ever, successful at reversing malignant obstruction. With this information, the patient decided to be discharged home with hospice and spend time with her family. She died peacefully at her home approximately two weeks later.

Article Outline

Futile Is as Futile Does

When deciding whether or not chemotherapy is “futile,” the concept of medical futility must be explored.[1] Though it remains difficult to adequately define, the qualitative and quantitative descriptions offered by Schneiderman et al[2] are widely used. Qualitatively, futile treatment “merely preserves permanent unconsciousness or cannot end dependence on intensive medical care.” More precisely, it is a medical treatment “that in the last 100 cases … has been useless.”[2] A useful, albeit imprecise, definition of futile chemotherapy is that in which the burdens and risks outweigh the benefits. As an example, studies on chemotherapy for advanced non-small-cell lung cancer (NSCLC) have shown that patients with poor performance status or chemotherapy-unresponsive disease receive little benefit in terms of response rates and survival. [3] and [4] A retrospective analysis by Massarelli et al3 showed dismal response rates for third- and fourth-line NSCLC chemotherapy of 2.3% and 0%, respectively. Additionally, an observational study by Zietemann and Duell[4] showed that 40% and 50% of patients receiving second- and third-line chemotherapy for NSCLC die during or soon after treatment, respectively, and that over 20% receive chemotherapy within 14 days of death. Neither study commented on quality of life experienced by patients. However, a recent study by Temel et al[5] demonstrated that NSCLC patients receiving concurrent palliative care and standard oncologic care had better quality of life and even longer survival than patients receiving only standard oncologic care, despite being less likely to receive aggressive end-of-life care. Though limited to patients with NSCLC, these studies illustrate that chemotherapy in advanced cancer is often futile, especially when less aggressive care can improve quality of life as well as survival.

 

 

Addressing the futility of chemotherapy with patients is challenging for most oncologists. Although defining treatments as “futile” is suitable in the medical literature, it is a word that may carry negative connotations, such as hopelessness or abandonment, to patients. A more descriptive and less negative term, “nonbeneficial,” may be used when discussing futile chemotherapy with patients. The point when chemotherapy becomes nonbeneficial, and thus futile, is different for each patient and might even change over time. Addressing the patient's definition of nonbeneficial chemotherapy regularly during treatment ensures that the patient's goals are clear and allows the oncologist to direct conversation toward alternative options, such as palliative and hospice care, when chemotherapy cannot provide the benefits sought by the patient. This can be as simple as asking the patient, “Do you think the chemotherapy is giving you enough benefit to continue?”

Palliative Care: It's Not Just Giving Up on People

Both the physician and the patient face several decisions when considering whether or not to pursue chemotherapy for advanced cancer. First of all, the patient must decide how much information he or she wants from the oncologist. If the patient is the decision maker, he or she must choose to accept chemotherapy that is palliative, not curative. After a frank discussion about the anticipated outcomes and symptoms associated with chemotherapy, the patient must consider whether he or she can accept the burden of treatment for the potential of prolonging life by days, weeks, or months. On the other hand, the oncologist must decide if chemotherapy should even be offered, based on patient performance status, known therapeutic outcomes, and patient values and goals. The oncologist can reassure patients that the best available data show that patients who use hospice for even one day actually live longer than those who do not.[6] Once informed about what palliative care and hospice offer, the patient may determine whether or not alternatives to chemotherapy are more favorable. If the patient qualifies for clinical trials, he or she must decide to accept treatment with uncertain outcome. When reflecting upon such difficult issues, both the patient and oncologist should involve others to help guide decision making. Oncologists can consult trusted colleagues for their expertise and to ensure that they are using the best information available. Patients should involve loved ones whom they trust to help make decisions in their best interest. Table 1 provides key questions that the oncologist faces when making these decisions and how to approach them.

Table 1: Questions to discuss with the patient when chemotherapy may be futile

Question

Leading prompts

Comment

What is the patient’s current understanding of the disease?

How much do you know about your cancer at this point?

 

 

How much do you want to know?

Be sure the patient is ready to discuss this issue and that you have enough time for discussion.

 

Ask if there are others who should receive this information simultaneously, afterwards, or instead of the patient.

What are the patient’s goals?

Knowing that we can’t cure your cancer, what are your goals, wishes, or hopes for the future?

Treatment decisions may be impacted greatly by a patient’s personal goals (e.g. patient wants to live to child’s graduation, or patient wants to be as comfortable as possible)

If chemotherapy is an option and the patient is interested, is he/she aware of potential risks and benefits?

Although everyone responds differently, these are the likely side effects and outcomes of this treatment…

Be specific in terms of likelihood of response, type of response (palliation instead of cure, extent of life prolongation expected, symptom relief, etc.) and how likely it is that treatment will help achieve patient’s goals.

 

Discuss potential symptom burden from treatment in detail.

 

Patient needs to be able to make informed decision about risks vs. benefits involved in potential treatment.

If the patient declines chemotherapy, treatment is not indicated, or treatment fails, what other options are available?

Let’s talk about options to make sure that you are comfortable and enjoy the highest quality of life possible in the time that you have left.

Focus on pain and symptom management. Discuss hospice options (home vs. inpatient) and make referrals when appropriate.

 

Stress that you will continue your relationship with the patient (possibly as their hospice provider) and that you will ensure that their symptoms are managed, either directly or through hospice nurses.

 

 

As an alternative to addressing the above issues with the patient independently, oncologists may involve a palliative care specialist to facilitate this conversation.[7] Particularly in cases where the oncologist decides that chemotherapy is no longer a viable option, it may be easier, from both the patient and the provider perspectives, for the palliative care specialist to have this discussion. In a recent survey of patients on our oncology ward, the great majority did not want to discuss advance directives (ADs) with their oncologist—these patients thought ADs were important and should be discussed but were more comfortable discussing them with the admitting provider than the oncologist.[8] Patients may feel that they are disappointing their oncologist by being unable to take further treatment or by admitting that treatment has failed them. Similarly, oncologists might view having this discussion as an admission of their failure as a provider. The palliative care specialist, on the other hand, has no responsibility for chemotherapy and possibly no prior relationship with the patient, thus alleviating this type of emotional association between provider and patient. Furthermore, the conversation about nonbeneficial chemotherapy provides a segue for the palliative care provider to discuss with patients what he or she does best: establishing goals of care, managing symptoms, and maintaining comfort. For the palliative care specialist, providing symptom management and the best possible quality of life for patients are the fundamental goals. Death is generally not viewed with a sense of failure when palliation is the focus of care.

Oncology: Palliative Care Is Giving Up

We still hear from oncologists like ourselves the dreaded words “What do you want me to do, give up on the patient?” or, to the patient, “What, are you giving up? I thought you'd keep fighting!” We would argue that current best practices include knowing when the risks and harms of chemotherapy outweigh any potential chance of benefit. Physicians and patients should follow current National Comprehensive Cancer Network (NCCN) guidelines for solid tumors such as breast9 and lung10 cancer and stop chemotherapy when the chance of success is minimal. If the doctor cannot describe a specific, substantial benefit that outweighs the toxicity, he or she should not recommend it.[11] And all the relevant guidelines call for considering a switch to nonchemotherapy palliative care when the patient's performance status is Eastern Cooperative Oncology Group (ECOG) ≥3, defined as “3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours.”[12] Such a simple threshold could dramatically reduce the use of chemotherapy at the end of life and lessen downstream toxicities.

Oncologists can implement several strategies to help facilitate the transition from aggressive care to comfort care (Table 2). For patients with incurable cancer, oncologists can hold early discussions about palliative and hospice options that will need to be implemented when chemotherapy is no longer able to control their disease. This discussion introduces palliative medicine as part of the care plan for incurable disease and allows the patient to anticipate such a transition. Oncologists can also provide reassurance that they will continue to be involved in their patient's care and to support them, even if the patient does not undergo further chemotherapy. There are at least four studies that show equal[13] or better[6] survival, smoother transitions to hospice when death is inevitable, less intensive end-of-life care, and superior patient and family outcomes with concurrent palliative care. [14] and [15] By helping patients establish legal documents, such as ADs and power of attorney, oncologists and palliative care specialists can alleviate some of the stress related to the end of life and make the transition to comfort care easier. Finally, oncologists can review guidelines such as those from the NCCN and American Society of Clinical Oncology, which call for a switch to palliative care when the cancer has grown on three regimens or the patient's ECOG performance status is three or above. [11] and [12]

Table 2: Things that help ONCOLOGISTS and their patients

Item

How it helps

Comments

Early discussion of palliative and hospice care when chemotherapy may no longer help.

Hospice (and eventual death) will not come as a complete surprise.

“We will do our best to help you with this cancer, but at some point there may not be any treatments known to help….” “Remember the conversation we had when we first met?...”

Reassurance that the oncologist will not abandon the patient if concurrent care is given.

This major fear may keep oncology patients at the same practice they have known for years – it is familiar – when they would be better served by transition.

There are now at least 4 randomized trials showing that most patients will accept concurrent palliative care if offered, and that outcomes are equal or better, at less cost.6,13,14,15

Legal documents such as Advance Medical Directives, Durable Medical Power of Attorney

Reinforces the seriousness and “now” aspect of care.

These are readily available in all states at no cost. They are not the final word on how to live one’s remaining time, but will get the conversation started.

Best nationally recognized information showing that further chemotherapy will not help due to 3 prior failures, or is not indicated due to poor performance status.9,10

The oncologist can point to the right page and say “The best national guidelines call for a switch away from chemo…because it will do no good and will cause harmful side effects.”

Readily accessed from the Internet.

Use decision aids, similar to Adjuvant!.

Increases the amount of truthful information given, even when the news is bad, and helps with transition points.

An increasing number of these are available[i],[ii],[iii],[iv] and will soon be offered as smart phone applications (aps).

 

 

Communication tools, such as the National Cancer Institute's Oncotalk and EPEC-O, are useful for oncologists seeking to further enhance their communication skills.

Take-Home Messages

Guiding patients in making decisions about nonbeneficial, or futile, chemotherapy presents a challenge for many oncologists as well as their patients and families. Though futility is difficult to define, oncologists and their patients can decide through regular, open discussion if the burdens of chemotherapy outweigh the benefits and whether or not chemotherapy can achieve the reasonable benefits desired by the patient. “Your cancer is advancing despite our best efforts to keep it from growing. Let's talk about what options we have at this point and see what will work best for you.” To make such decisions, oncologists must obtain the most current information and convey it to patients (or their designated decision makers) as clearly as possible. “Based on the latest evidence, there is a 20% chance that the cancer will shrink or stay the same size with this treatment and an 80% chance that it will continue to grow despite treatment.” Both oncologists and their patients should involve those whom they trust to help with decision making. In cases where chemotherapy is nonbeneficial, oncologists may prefer to involve palliative and hospice care specialists to discuss the transition to comfort care with the patient. “At this time, I do not have any treatments that are likely to help you live longer or more comfortably, but I want to make sure that we get the most out of the rest of your life. I have asked a palliative care specialist to help us make this possible.” In order to ease the transition from aggressive or curative care to comfort care, oncologists can employ approaches such as early discussion of palliative and hospice care, assuring the patient of continued involvement in their care, and helping patients with ADs. These approaches not only benefit patients and their families but also strengthen the relationship between the oncologist and the patients and their families.

Acknowledgments

This research was supported by grants GO8 LM0095259 from the National Library of Medicine and R01CA116227-01 (both to T. J. S.) from the National Cancer Institute.

References [PubMed ID in brackets]

1 P.R. Helft, M. Siegler and J. Lantos, The rise and fall of the medical futility movement, . N Engl J Med,  343  (2000), pp. 293–296 [10911014].

2 L.J. Schneiderman, N.S. Jecker and A.R. Jonsen, Medical futility: its meaning and ethical implications, . Ann Intern Med,  112  (1990), pp. 949–954 [2187394].

3 E. Massarelli, F. Andre, D.D. Liu, J.J. Lee, M. Wolf, A. Fandi, J. Ochs, T. Le Chevalier, F. Fossella and R.S. Herbst, A retrospective analysis of the outcome of patients who have received two prior chemotherapy regimens including platinum and docetaxel for recurrent non-small-cell lung cancer, . Lung Cancer,  39 1 (2003), pp. 55–61 [12499095].

4 V. Zietemann and T. Duell, Every-day clinical practice in patients with advanced non-small-cell lung cancer, . Lung Cancer,  68 2 (2010), pp. 273–277 [19632737].

5 J.S. Temel, J.A. Greer, A. Muzikansky, E.R. Gallagher, S. Admane, V.A. Jackson, C.M. Dahlin, C.D. Blinderman, J. Jacobsen, W.F. Pirl, J.A. Billings and T.J. Lynch, Early palliative care for patients with metastatic non-small-cell lung cancer, . N Engl J Med,  363  (2010), pp. 733–742 [20818875].

6 S.R. Connor, B. Pyenson, K. Fitch, C. Spence and K. Iwasaki, Comparing hospice and nonhospice patient survival among patients who die within a three-year window. J Pain Symptom Manage,  33 3 (2007), pp. 238–246.

7 S.E. Harrington and T.J. Smith, The role of chemotherapy at the end of life: “when is enough, enough?”. JAMA,  299 22 (2008), pp. 2667–2678.

8 L. Dow, R. Matsuyama, L. Kuhn, L. Lyckholm, E.B. Lamont and T.J. Smith, Paradoxes in advance care planning. J Clin Oncol,  28 2 (2010), pp. 299–304.

9 NCCN Breast Cancer Clinical Practice Guidelines Panel, R.W. Carlson, D.C. Allred, B.O. Anderson, H.J. Burstein, W.B. Carter, S.B. Edge, J.K. Erban, W.B. Farrar, L.J. Goldstein, W.J. Gradishar, D.F. Hayes, C.A. Hudis, M. Jahanzeb, K. Kiel, B.M. Ljung, P.K. Marcom, I.A. Mayer, B. McCormick, L.M. Nabell, L.J. Pierce, E.C. Reed, M.L. Smith, G. Somlo, R.L. Theriault, N.S. Topham, J.H. Ward, E.P. Winer and A.C. Wolff, Breast cancer. J Natl Compr Canc Netw,  7 2 (2009), pp. 122–192.

10 NCCN Non-Small Cell Lung Cancer Panel Members, D.S. Ettinger, W. Akerley, G. Bepler, M.G. Blum, A. Chang, R.T. Cheney, L.R. Chirieac, T.A. D'Amico, T.L. Demmy, A.K. Ganti, R. Govindan, F.W. Grannis, T. Jahan, M. Jahanzeb, D.H. Johnson, A. Kessinger, R. Komaki, F.M. Kong, M.G. Kris, L.M. Krug, Q.T. Le, I.T. Lennes, R. Martins, J. O'Malley, R.U. Osarogiagbon, G.A. Otterson, J.D. Patel, K.M. Pisters, K. Reckamp, G.J. Riely, E. Rohren, G.R. Simon, S.J. Swanson, D.E. Wood and S.C. Yang, Non-small cell lung cancer. J Natl Compr Canc Netw,  8 7 (2010), pp. 740–801.

 

 

11 American Society of Clinical Oncology Outcomes Working Group, Outcomes of cancer treatment for technology assessment and cancer treatment guidelines. J Clin Oncol,  14  (1996), pp. 671–679.

12 Eastern Cooperative Oncology Group, ECOG Performance Status, http://ecog.dfci.harvard.edu/general/perf_stat.html Accessed November 30, 2010. 

13 J. Finn, K. Pienta and J. Parzuchowski, Bridging cancer treatment and hospice care. Proc Am Soc Clin Oncol,  21  (2002), p. 1452. 

14 G. Gade, I. Venohr, D. Conner, K. McGrady, J. Beane, R.H. Richardson, M.P. Williams, M. Liberson, M. Blum and R. Della Penna, Impact of an inpatient palliative care team: a randomized control trial. J Palliat Med,  11 2 (2008), pp. 180–190.

Copyright © 2011 Elsevier Inc. All rights reserved.


The Journal of Supportive Oncology
Volume 9, Issue 5, September-October 2011, Pages 184-187


doi:10.1016/j.suponc.2011.04.001 
  Permissions & Reprints

How We Do It

Guiding Patients Facing Decisions about “Futile” Chemotherapy

Erin Alesi MD, Barton Bobb RN, MSN, Thomas J. Smith MD 

Received 4 April 2011; Accepted 16 December 2011. Available online 24 September 2011.

Case Presentation

Ms. G is a 71-year-old woman with metastatic gastric adenocarcinoma recently diagnosed after an extensive surgical resection for a small bowel obstruction (SBO). She was admitted from the surgery clinic with intractable nausea and vomiting. An abdominal computerized tomographic (CT) scan revealed a partial SBO and peritoneal carcinomatosis. Given her recent surgery, the extent of her disease, and high likelihood of recurrent SBO, the surgical team decided that Ms. G was no longer a surgical candidate. When her symptoms did not improve with conservative measures, both oncology and palliative medicine were consulted to assist with symptom management and goals of care. The oncology team stated that Ms. G was still a chemotherapy candidate and suggested that she attend her new patient evaluation in oncology clinic the following week. The palliative medicine team then met with the patient to discuss management options and her preferences for care. The palliative care team explained ways to control her nausea and vomiting without using a nasogastric tube, and the patient agreed to transfer to their service for symptom management. The palliative team explained that her cancer was incurable but that chemotherapy options existed to help control her disease and possibly prolong her life. They also explained that the chemotherapy has side effects and that the patient would need to decide if she wanted to undergo treatment and accept potential side effects for the possibility of prolonging her life by weeks to months and improving her symptoms. As an alternative, she was told that she could focus solely on symptom control with medications and allow her disease to take its natural course. Ms. G was asked to think about how she wanted to spend the time she had left. Prior to discharge, as her symptoms improved, Ms. G was evaluated by another oncologist, who, after consulting the expert gastrointestinal cancer team, explained to her that the current chemotherapy options available for metastatic gastric cancer were rarely, if ever, successful at reversing malignant obstruction. With this information, the patient decided to be discharged home with hospice and spend time with her family. She died peacefully at her home approximately two weeks later.

Article Outline

Futile Is as Futile Does

When deciding whether or not chemotherapy is “futile,” the concept of medical futility must be explored.[1] Though it remains difficult to adequately define, the qualitative and quantitative descriptions offered by Schneiderman et al[2] are widely used. Qualitatively, futile treatment “merely preserves permanent unconsciousness or cannot end dependence on intensive medical care.” More precisely, it is a medical treatment “that in the last 100 cases … has been useless.”[2] A useful, albeit imprecise, definition of futile chemotherapy is that in which the burdens and risks outweigh the benefits. As an example, studies on chemotherapy for advanced non-small-cell lung cancer (NSCLC) have shown that patients with poor performance status or chemotherapy-unresponsive disease receive little benefit in terms of response rates and survival. [3] and [4] A retrospective analysis by Massarelli et al3 showed dismal response rates for third- and fourth-line NSCLC chemotherapy of 2.3% and 0%, respectively. Additionally, an observational study by Zietemann and Duell[4] showed that 40% and 50% of patients receiving second- and third-line chemotherapy for NSCLC die during or soon after treatment, respectively, and that over 20% receive chemotherapy within 14 days of death. Neither study commented on quality of life experienced by patients. However, a recent study by Temel et al[5] demonstrated that NSCLC patients receiving concurrent palliative care and standard oncologic care had better quality of life and even longer survival than patients receiving only standard oncologic care, despite being less likely to receive aggressive end-of-life care. Though limited to patients with NSCLC, these studies illustrate that chemotherapy in advanced cancer is often futile, especially when less aggressive care can improve quality of life as well as survival.

 

 

Addressing the futility of chemotherapy with patients is challenging for most oncologists. Although defining treatments as “futile” is suitable in the medical literature, it is a word that may carry negative connotations, such as hopelessness or abandonment, to patients. A more descriptive and less negative term, “nonbeneficial,” may be used when discussing futile chemotherapy with patients. The point when chemotherapy becomes nonbeneficial, and thus futile, is different for each patient and might even change over time. Addressing the patient's definition of nonbeneficial chemotherapy regularly during treatment ensures that the patient's goals are clear and allows the oncologist to direct conversation toward alternative options, such as palliative and hospice care, when chemotherapy cannot provide the benefits sought by the patient. This can be as simple as asking the patient, “Do you think the chemotherapy is giving you enough benefit to continue?”

Palliative Care: It's Not Just Giving Up on People

Both the physician and the patient face several decisions when considering whether or not to pursue chemotherapy for advanced cancer. First of all, the patient must decide how much information he or she wants from the oncologist. If the patient is the decision maker, he or she must choose to accept chemotherapy that is palliative, not curative. After a frank discussion about the anticipated outcomes and symptoms associated with chemotherapy, the patient must consider whether he or she can accept the burden of treatment for the potential of prolonging life by days, weeks, or months. On the other hand, the oncologist must decide if chemotherapy should even be offered, based on patient performance status, known therapeutic outcomes, and patient values and goals. The oncologist can reassure patients that the best available data show that patients who use hospice for even one day actually live longer than those who do not.[6] Once informed about what palliative care and hospice offer, the patient may determine whether or not alternatives to chemotherapy are more favorable. If the patient qualifies for clinical trials, he or she must decide to accept treatment with uncertain outcome. When reflecting upon such difficult issues, both the patient and oncologist should involve others to help guide decision making. Oncologists can consult trusted colleagues for their expertise and to ensure that they are using the best information available. Patients should involve loved ones whom they trust to help make decisions in their best interest. Table 1 provides key questions that the oncologist faces when making these decisions and how to approach them.

Table 1: Questions to discuss with the patient when chemotherapy may be futile

Question

Leading prompts

Comment

What is the patient’s current understanding of the disease?

How much do you know about your cancer at this point?

 

 

How much do you want to know?

Be sure the patient is ready to discuss this issue and that you have enough time for discussion.

 

Ask if there are others who should receive this information simultaneously, afterwards, or instead of the patient.

What are the patient’s goals?

Knowing that we can’t cure your cancer, what are your goals, wishes, or hopes for the future?

Treatment decisions may be impacted greatly by a patient’s personal goals (e.g. patient wants to live to child’s graduation, or patient wants to be as comfortable as possible)

If chemotherapy is an option and the patient is interested, is he/she aware of potential risks and benefits?

Although everyone responds differently, these are the likely side effects and outcomes of this treatment…

Be specific in terms of likelihood of response, type of response (palliation instead of cure, extent of life prolongation expected, symptom relief, etc.) and how likely it is that treatment will help achieve patient’s goals.

 

Discuss potential symptom burden from treatment in detail.

 

Patient needs to be able to make informed decision about risks vs. benefits involved in potential treatment.

If the patient declines chemotherapy, treatment is not indicated, or treatment fails, what other options are available?

Let’s talk about options to make sure that you are comfortable and enjoy the highest quality of life possible in the time that you have left.

Focus on pain and symptom management. Discuss hospice options (home vs. inpatient) and make referrals when appropriate.

 

Stress that you will continue your relationship with the patient (possibly as their hospice provider) and that you will ensure that their symptoms are managed, either directly or through hospice nurses.

 

 

As an alternative to addressing the above issues with the patient independently, oncologists may involve a palliative care specialist to facilitate this conversation.[7] Particularly in cases where the oncologist decides that chemotherapy is no longer a viable option, it may be easier, from both the patient and the provider perspectives, for the palliative care specialist to have this discussion. In a recent survey of patients on our oncology ward, the great majority did not want to discuss advance directives (ADs) with their oncologist—these patients thought ADs were important and should be discussed but were more comfortable discussing them with the admitting provider than the oncologist.[8] Patients may feel that they are disappointing their oncologist by being unable to take further treatment or by admitting that treatment has failed them. Similarly, oncologists might view having this discussion as an admission of their failure as a provider. The palliative care specialist, on the other hand, has no responsibility for chemotherapy and possibly no prior relationship with the patient, thus alleviating this type of emotional association between provider and patient. Furthermore, the conversation about nonbeneficial chemotherapy provides a segue for the palliative care provider to discuss with patients what he or she does best: establishing goals of care, managing symptoms, and maintaining comfort. For the palliative care specialist, providing symptom management and the best possible quality of life for patients are the fundamental goals. Death is generally not viewed with a sense of failure when palliation is the focus of care.

Oncology: Palliative Care Is Giving Up

We still hear from oncologists like ourselves the dreaded words “What do you want me to do, give up on the patient?” or, to the patient, “What, are you giving up? I thought you'd keep fighting!” We would argue that current best practices include knowing when the risks and harms of chemotherapy outweigh any potential chance of benefit. Physicians and patients should follow current National Comprehensive Cancer Network (NCCN) guidelines for solid tumors such as breast9 and lung10 cancer and stop chemotherapy when the chance of success is minimal. If the doctor cannot describe a specific, substantial benefit that outweighs the toxicity, he or she should not recommend it.[11] And all the relevant guidelines call for considering a switch to nonchemotherapy palliative care when the patient's performance status is Eastern Cooperative Oncology Group (ECOG) ≥3, defined as “3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours.”[12] Such a simple threshold could dramatically reduce the use of chemotherapy at the end of life and lessen downstream toxicities.

Oncologists can implement several strategies to help facilitate the transition from aggressive care to comfort care (Table 2). For patients with incurable cancer, oncologists can hold early discussions about palliative and hospice options that will need to be implemented when chemotherapy is no longer able to control their disease. This discussion introduces palliative medicine as part of the care plan for incurable disease and allows the patient to anticipate such a transition. Oncologists can also provide reassurance that they will continue to be involved in their patient's care and to support them, even if the patient does not undergo further chemotherapy. There are at least four studies that show equal[13] or better[6] survival, smoother transitions to hospice when death is inevitable, less intensive end-of-life care, and superior patient and family outcomes with concurrent palliative care. [14] and [15] By helping patients establish legal documents, such as ADs and power of attorney, oncologists and palliative care specialists can alleviate some of the stress related to the end of life and make the transition to comfort care easier. Finally, oncologists can review guidelines such as those from the NCCN and American Society of Clinical Oncology, which call for a switch to palliative care when the cancer has grown on three regimens or the patient's ECOG performance status is three or above. [11] and [12]

Table 2: Things that help ONCOLOGISTS and their patients

Item

How it helps

Comments

Early discussion of palliative and hospice care when chemotherapy may no longer help.

Hospice (and eventual death) will not come as a complete surprise.

“We will do our best to help you with this cancer, but at some point there may not be any treatments known to help….” “Remember the conversation we had when we first met?...”

Reassurance that the oncologist will not abandon the patient if concurrent care is given.

This major fear may keep oncology patients at the same practice they have known for years – it is familiar – when they would be better served by transition.

There are now at least 4 randomized trials showing that most patients will accept concurrent palliative care if offered, and that outcomes are equal or better, at less cost.6,13,14,15

Legal documents such as Advance Medical Directives, Durable Medical Power of Attorney

Reinforces the seriousness and “now” aspect of care.

These are readily available in all states at no cost. They are not the final word on how to live one’s remaining time, but will get the conversation started.

Best nationally recognized information showing that further chemotherapy will not help due to 3 prior failures, or is not indicated due to poor performance status.9,10

The oncologist can point to the right page and say “The best national guidelines call for a switch away from chemo…because it will do no good and will cause harmful side effects.”

Readily accessed from the Internet.

Use decision aids, similar to Adjuvant!.

Increases the amount of truthful information given, even when the news is bad, and helps with transition points.

An increasing number of these are available[i],[ii],[iii],[iv] and will soon be offered as smart phone applications (aps).

 

 

Communication tools, such as the National Cancer Institute's Oncotalk and EPEC-O, are useful for oncologists seeking to further enhance their communication skills.

Take-Home Messages

Guiding patients in making decisions about nonbeneficial, or futile, chemotherapy presents a challenge for many oncologists as well as their patients and families. Though futility is difficult to define, oncologists and their patients can decide through regular, open discussion if the burdens of chemotherapy outweigh the benefits and whether or not chemotherapy can achieve the reasonable benefits desired by the patient. “Your cancer is advancing despite our best efforts to keep it from growing. Let's talk about what options we have at this point and see what will work best for you.” To make such decisions, oncologists must obtain the most current information and convey it to patients (or their designated decision makers) as clearly as possible. “Based on the latest evidence, there is a 20% chance that the cancer will shrink or stay the same size with this treatment and an 80% chance that it will continue to grow despite treatment.” Both oncologists and their patients should involve those whom they trust to help with decision making. In cases where chemotherapy is nonbeneficial, oncologists may prefer to involve palliative and hospice care specialists to discuss the transition to comfort care with the patient. “At this time, I do not have any treatments that are likely to help you live longer or more comfortably, but I want to make sure that we get the most out of the rest of your life. I have asked a palliative care specialist to help us make this possible.” In order to ease the transition from aggressive or curative care to comfort care, oncologists can employ approaches such as early discussion of palliative and hospice care, assuring the patient of continued involvement in their care, and helping patients with ADs. These approaches not only benefit patients and their families but also strengthen the relationship between the oncologist and the patients and their families.

Acknowledgments

This research was supported by grants GO8 LM0095259 from the National Library of Medicine and R01CA116227-01 (both to T. J. S.) from the National Cancer Institute.

References [PubMed ID in brackets]

1 P.R. Helft, M. Siegler and J. Lantos, The rise and fall of the medical futility movement, . N Engl J Med,  343  (2000), pp. 293–296 [10911014].

2 L.J. Schneiderman, N.S. Jecker and A.R. Jonsen, Medical futility: its meaning and ethical implications, . Ann Intern Med,  112  (1990), pp. 949–954 [2187394].

3 E. Massarelli, F. Andre, D.D. Liu, J.J. Lee, M. Wolf, A. Fandi, J. Ochs, T. Le Chevalier, F. Fossella and R.S. Herbst, A retrospective analysis of the outcome of patients who have received two prior chemotherapy regimens including platinum and docetaxel for recurrent non-small-cell lung cancer, . Lung Cancer,  39 1 (2003), pp. 55–61 [12499095].

4 V. Zietemann and T. Duell, Every-day clinical practice in patients with advanced non-small-cell lung cancer, . Lung Cancer,  68 2 (2010), pp. 273–277 [19632737].

5 J.S. Temel, J.A. Greer, A. Muzikansky, E.R. Gallagher, S. Admane, V.A. Jackson, C.M. Dahlin, C.D. Blinderman, J. Jacobsen, W.F. Pirl, J.A. Billings and T.J. Lynch, Early palliative care for patients with metastatic non-small-cell lung cancer, . N Engl J Med,  363  (2010), pp. 733–742 [20818875].

6 S.R. Connor, B. Pyenson, K. Fitch, C. Spence and K. Iwasaki, Comparing hospice and nonhospice patient survival among patients who die within a three-year window. J Pain Symptom Manage,  33 3 (2007), pp. 238–246.

7 S.E. Harrington and T.J. Smith, The role of chemotherapy at the end of life: “when is enough, enough?”. JAMA,  299 22 (2008), pp. 2667–2678.

8 L. Dow, R. Matsuyama, L. Kuhn, L. Lyckholm, E.B. Lamont and T.J. Smith, Paradoxes in advance care planning. J Clin Oncol,  28 2 (2010), pp. 299–304.

9 NCCN Breast Cancer Clinical Practice Guidelines Panel, R.W. Carlson, D.C. Allred, B.O. Anderson, H.J. Burstein, W.B. Carter, S.B. Edge, J.K. Erban, W.B. Farrar, L.J. Goldstein, W.J. Gradishar, D.F. Hayes, C.A. Hudis, M. Jahanzeb, K. Kiel, B.M. Ljung, P.K. Marcom, I.A. Mayer, B. McCormick, L.M. Nabell, L.J. Pierce, E.C. Reed, M.L. Smith, G. Somlo, R.L. Theriault, N.S. Topham, J.H. Ward, E.P. Winer and A.C. Wolff, Breast cancer. J Natl Compr Canc Netw,  7 2 (2009), pp. 122–192.

10 NCCN Non-Small Cell Lung Cancer Panel Members, D.S. Ettinger, W. Akerley, G. Bepler, M.G. Blum, A. Chang, R.T. Cheney, L.R. Chirieac, T.A. D'Amico, T.L. Demmy, A.K. Ganti, R. Govindan, F.W. Grannis, T. Jahan, M. Jahanzeb, D.H. Johnson, A. Kessinger, R. Komaki, F.M. Kong, M.G. Kris, L.M. Krug, Q.T. Le, I.T. Lennes, R. Martins, J. O'Malley, R.U. Osarogiagbon, G.A. Otterson, J.D. Patel, K.M. Pisters, K. Reckamp, G.J. Riely, E. Rohren, G.R. Simon, S.J. Swanson, D.E. Wood and S.C. Yang, Non-small cell lung cancer. J Natl Compr Canc Netw,  8 7 (2010), pp. 740–801.

 

 

11 American Society of Clinical Oncology Outcomes Working Group, Outcomes of cancer treatment for technology assessment and cancer treatment guidelines. J Clin Oncol,  14  (1996), pp. 671–679.

12 Eastern Cooperative Oncology Group, ECOG Performance Status, http://ecog.dfci.harvard.edu/general/perf_stat.html Accessed November 30, 2010. 

13 J. Finn, K. Pienta and J. Parzuchowski, Bridging cancer treatment and hospice care. Proc Am Soc Clin Oncol,  21  (2002), p. 1452. 

14 G. Gade, I. Venohr, D. Conner, K. McGrady, J. Beane, R.H. Richardson, M.P. Williams, M. Liberson, M. Blum and R. Della Penna, Impact of an inpatient palliative care team: a randomized control trial. J Palliat Med,  11 2 (2008), pp. 180–190.

Copyright © 2011 Elsevier Inc. All rights reserved.


Publications
Publications
Topics
Article Type
Display Headline
Guiding Patients Facing Decisions about “Futile” Chemotherapy
Display Headline
Guiding Patients Facing Decisions about “Futile” Chemotherapy
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Supporting Children's Grief within an Adult and Pediatric Palliative Care Program

Article Type
Changed
Fri, 01/04/2019 - 11:03
Display Headline
Supporting Children's Grief within an Adult and Pediatric Palliative Care Program

Children are too often the forgotten mourners in the homes of dying patients. Children, even young children, and youth grieve and mourn the threatened and, then, actual loss of a dying parent, sibling, or other significant family member.1 At a time when the family resources and focus are pulled away and taxed, caregivers are tasked with the difficult job of sorting through their own emotions and a wealth of advice. Caregivers must decide how they will communicate with, include, and support the children/youth in their care.

Although evidence is incomplete and there is a clear need for further studies, links between unresolved childhood grief, or an inability to adequately process their grief, and subsequent psychiatric conditions such as depression and anxiety have been presented as far back as Freud.[2], [3], [4] and [5] In addition, prevalent feelings of responsibility and exclusion and poor communication are consistently identified by researchers interviewing bereaved children/youth about their own experience over the last couple of decades.[6], [7] and [8] Therefore, given the risk of negative psychological and social outcomes associated with children's grief and the struggles communicated by children themselves, it is critical to recognize the important and preventive role of supportive interventions, especially prior to the death of a significant family member.

Looking at caregivers' experiences, there is still a large divide between the advice given by many family and friends in this situation (see Table 1) and what has become accepted within the palliative and grief counseling fields as “best practice.”[6], [7] and [8] In addition, family members' access to professionals trained or knowledgeable in this area is growing but usually still limited.9 Many children/youth are left uninformed, unprepared, and cut off from their family's support.

Table 1. Myths and Realities about Speaking to Children about Grief and Dying. Adapted from MacPherson C.10

 

 

Professionals are not immune to subscribing to the myths listed in Table 1 and “are often inhibited by their anxieties about saying or doing the wrong thing and causing lasting emotional damage.”10 However, by communicating openly and honestly and including children/youth, informed care team members can offer many supportive interventions that a family can benefit from during the time leading up to and following the death. These interventions foster the best outcomes when they are offered early on in the palliative trajectory.[11] and [12]

Our Setting

The Temmy Latner Centre for Palliative Care (TLCPC) at Mount Sinai Hospital in Toronto is one of Canada's largest academic palliative care programs, incorporating a children's center, the Max and Beatrice Wolfe Children's Centre, which provides pediatric palliative care and children's grief programs (Dr. Jay Children's Grief Program). Our children's center supports children, youth, and their families when a family member is dying or has died. This support includes Canada's first Camp Erin, an overnight children's grief camp. Children are referred to these programs for grief counseling by our center's palliative care physicians, local palliative care units and hospices, and a wide variety of community agencies. Children's grief programs are very limited in our large urban setting, as is true in most communities across North America. We have four counselors devoted to child and youth grief support services.

What We Do

Action 1: Intake and Assessment

The center has an open referral policy, accepting referrals from any source, including self-referrals, regardless of the nature of the illness or cause of death.

An intake phone call, lasting between 15 and 60 minutes, is made to the family to assess their needs and to provide psychoeducation and relevant resources. Based on our belief that early intervention provides the most supportive opportunities, families in which the patient is dying are prioritized.

As a result of demand for services being greater than our resources, children and their families bereaved at the time of referral are provided with an initial psychoeducational visit and assessment and then placed on a waiting list if further counseling is deemed appropriate.

In either case, the initial assessment often reveals that what the family needs most is psychoeducation about child/youth grief, communication and development, and reassurance about the benefits of the things they are already doing. Caregivers are provided with educational materials, including a copy of the center's publication Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers,11 a list of Web resources, books, and brochures written by us. All these resources are also available to professionals.

Action 2: Counseling

If further assessment and counseling are warranted, children are seen individually, with siblings and/or with their family depending on the needs and circumstances. The bulk of our counseling services are brief, typically lasting three or four sessions in total. However, more intensive counseling is available on a case-by-case basis, with progress and needs being assessed every four sessions. Counseling techniques including expressive arts, crafts, therapeutic play, and activities are used to support children/youth and families in the grieving process. Families are able to contact counselors when issues arise for them, which often occurs around anniversaries, holidays, other important events, and as children/youth develop and experience their grief in a new light.13

The center's model is resiliency-based, nonpathological, and family-centered. Caregivers are empowered as primary and ongoing sources of support and dominant role models for the grieving child and youth in their care. In addition to conversations with their counselor, families are offered monthly opportunities forcaregiver education and peer support and various therapeutic group activities for children/youth.

Action 3: Complex Cases

Referrals to secondary children's mental health services are facilitated for families with needs beyond the scope of our supportive grief services, including children with indicators of complicated grief who need more intensive counseling. For children with severe psychological distress, referrals are made to a pediatric psychiatrist with special interest in this area.

According to Rando,14 there are a number of forms of unresolved or complicated grief which can overlap, and each has components of denial or regression. These include feelings of grief and mourning being absent, an inhibition of some of the normal symptoms of grief, putting grief on hold for any reason, and when there is a dependent or ambivalent relationship with the deceased. Two common manifestations are extreme anger and extreme guilt.

Some types of death that place children/youth at risk for complicated grief include a sudden or unexpected death, a violent death, a death involving mutilation, the death of a child, and death as the result of a prolonged illness. Also included is complicated loss associated with social stigma such as imprisonment, suicide, AIDS, abortion, severe mental illness, serious family dysfunction, or addiction. However, the presence of these factors does not necessarily lead to complicated grief. Complicated loss is known to be mediated by personal, familial, and social factors that contribute to relative risk and resilience.

What We Say

Engaging a family early in the palliative journey allows greater opportunity to prepare children/youth and prevent possible negative outcomes.[6] and [15] Christ and Weisenfluh16 tell us that the greatest need for support is found during the weeks leading up to the death. A large component of the early intervention we offer is age-appropriate psychoeducation to help caregivers conceptualize how their child/youth may be experiencing and understanding what is happening. Caregivers who anticipate some of the thoughts, feelings, questions, and struggles that their child/youth might face are empowered and children benefit.

The 3 Cs

1. Can I catch it?
2. Did I cause it?
3. Who is going to take care of me?

Julie Stokes15 was able to summarize what children/youth think and worry about most when a family member is dying into three questions. We have coined these three questions the “Three Cs”: Catch, Cause, and Care.

Catch

The first “C” relates to the fear expressed by children/youth that they could catch the illness. Melanie, a 7-year-old, explained that she would have a brain tumor soon because her sister Sarah, who was dying of brain cancer at the time, “is my sister and we lived in the same room always.” If such concerns go unexplored and children are not given clear information, caregivers may see children/youth distance themselves from their ill family member, develop a fear that they and the rest of their family will get sick and die as well, and other implications.17

The weight of language in a child's understanding of illness, death, and dying cannot be emphasized enough.11 More often than not, indirect and generic language used by adults, such as referring to someone as being “sick” or “not doing well,” complicates a child's ability to differentiate between the common cold or flu and life-threatening diseases and illnesses. “My mom was sick and she died; therefore, all people that are sick die.” Many adults believe that their child is too young to understand what cancer means. While it is true that children may not be able to grasp the complex medical information about the illness, they are able to understand a great deal more than they are given credit for, and using the word “cancer” gives them a way to distinguish their dying family member's illness from others such as the common cold or flu.

Cause

The second “C” arises from the common thought in children/youth that they somehow caused or hastened the death and/or prevented the recovery of their family member. Cause is one of the five accepted subconcepts of the developmental understanding of death8 and among the last to be mastered due to its complexity and abstract components. As she sat under her pink bunk-bed, 5-year-old Tayah told her counselor that she had cancer when she was a little girl and then her mother got it after her. Eleven-year-old Joshua shared, “I just have to get to that cancer walk thing. If I can do that, then my mom will get her cure. That is what they said on TV you know, to walk for the cure!”

There are a number of ways that children/youth may try to own responsibility for the illness and death of their family member. They may believe that things they thought or did not think, did or did not do, felt or did not feel were directly related to the cause, progression, or death of the family member.18 These thoughts are especially strong for children in the magical thinking stage of development. In Piaget's theory of cognitive development, magical thinking dominates the preoperational stage (2–7 years old) and describes thinking disconnected from the laws of nature.19 Special attention should be given to assuring children in this age group that they are in no way responsible for the illness or death of their family member, as well as to avoid minimizing these fears and beliefs of responsibility.[6] and [17] This point will need to be readdressed often, with frequent reassurance that they are not responsible.

Care

The third “C” includes concerns about what will become of them and who will take care of them as well as the desire to help care for the dying person. While it is commonly thought that this fear is felt more significantly by children/youth who have a dying or deceased parent, siblings of dying or deceased children appear to be just as challenged. They will experience the same break in their belief that their family member will always be with them.20 Also, many children worry that their surviving parent will be incapacitated and unable to care for them after the death. Four-year-old Alex looked up at his mom one night and said, “You are dying in front of me. I'm scared. Who is going to turn out the light, I can't reach it, and who will cook for me, I can't turn on the stove?” This fear extends beyond who will take over the practical parental roles, to the worry that the child or youth will be orphaned.

Children will often connect their own experience to what they have seen or heard and fear the worst.[18] and [21] Orphans are ever present in children's literature and movies and are often depicted as abandoned to fend for themselves. For many, this fear is grounded in the reality that their current caregiver may not have been emotionally able to address issues of custody or guardianship or that they do not have anyone willing or able to care for the kids after their death. As Alice lay on her bedroom floor writing notes to her dying mother in the critical care unit, she shared, “My mommy and my daddy are dying … they is both going to die,” convinced that this was the truth. Alice's dad is healthy and has been for all of Alice's life.

“Three Cs” and Adolescents

Occasionally, adults have questioned the relativity of the “Three Cs” to our adolescent (ages 13–18) population. Our clinical work has led us to conclude that indeed the “Three Cs” are very real in the lives of the youth we work with. An example of this in relation to “Catch” is Sarah, who expressed distress over the familial traits of breast cancer and the likelihood that her fate will one day mirror that of her mother, aunt, and grandmother.

With regard to “Cause,” we hear youth talk about the relationship between the quality of their caregiving efforts and the death of their family member. Many youth express a feeling that if they had done a better job caregiving, their family member would not have died, especially for youth in primary caregiving roles and single-parent homes.14

When looking at “Care,” youth are impacted by their ability to think abstractly and to experience the loss of what is yet to come. Youth understand that they depend on their caregivers for much more than their practical needs. Joshua talked to his counselor about the loss he was feeling as he searched for employment without his father's help and network. He shared, “If my Dad was here, he would know someone; he would know what I should do.” Important to note is that age is not always the best predictor of cognitive capacity as we see school-aged children grapple with many of the thoughts more commonly expressed by our older population.

Whether or not a caregiver has already spoken with a child about a diagnosis and prognosis, it is helpful to get him or her to consider what the child may be thinking about. Using the foundation of the “Three Cs” will help caregivers understand the importance of open, honest, and concrete communication, as well as feel better equipped to provide their child/youth with support.

Teaching Parents How to Communicate with Their Children

It is difficult for a parent to witness the reaction of a child/youth to such difficult information. This challenge cannot be questioned. However, the benefit to children/youth of having their parents lead or participate in communications and psychoeducation about the cancer and terminal prognosis is clear.[6], [7], [8] and [12] Caregivers are able to clarify assumptions and misperceptions while modeling that it is permissible to talk about cancer and dying together.

Medical staff can facilitate opportunities for such family communications by including children in family meetings and discussions with medical staff and by explaining complex concepts to family members in a simple, concrete fashion. Caregivers often need professionals to welcome and encourage the children to be included. Staff can reassure parents that although they may fear that their children's inclusion will be harmful, on the contrary, inclusion is helpful.

Getting Started

1 Ask what the child/youth understands about the disease/illness.
2 Fill in any gaps in their understanding and explain the treatments that were given.
3 Explain cancer treatments such as chemotherapy, radiation, and surgery.
4 Ask the child/youth what he or she thinks is going to happen.
5 Explain in terms that can be understood that the family member will die.

In any conversation with children/youth about such complex and consequential material, follow their lead in regard to the amount of detail they want. Conceptualize it as an onion: You will want to provide the child/youth with clear and simple language describing basic information. Beyond that, peel off subsequent layers as the child/youth requests more information, always using clear and concrete language. The child/youth may be uncomfortable with the material and may attempt to change the topic. This is a healthy coping mechanism; the average child is quite good at knowing when he or she has reached the limit of their emotional attention span. Allow the child to take the conversation in a different direction, periodically checking to see if he or she is ready to come back to the topic at hand. The benefit of starting this course of conversation early is that it allows children/youth the chance to receive information in small, digestible pieces and the time to process and integrate the information to begin to make meaning of it.

Summary

“What about the kids?” is a dominant and consuming question for caregivers supporting children/youth around the dying and death of a family member. The concerns and fears encompassed in this question can overwhelm caregivers as they put vast amounts of energy into trying to protect children/youth from the suffering and pain that awaits them. Perhaps the hardest lesson these caregivers must learn is that they cannot protect their child/youth from the death any more than they can stop the death from happening. Instead, what is needed most from children/youth is to be included, prepared, and provided with a safe place for emotional expression. Children, as well as adults, will grieve in their own specific way, mediated by their developmental level, circumstances of the illness and death, and protective factors available to them. Providing comprehensive, whole-person care to palliative patients with children/youth in their care ought to include psychoeducation and support for all members of the family. Our communities' bereaved children/youth will be impacted by the death of their family members in countless ways throughout their lives. As professionals caring for people who are dying, we have a responsibility to mediate this impact to the best of our ability.

 

 

References1

1 J. Bowlby, Pathological mourning and childhood mourning, J Am Psychoanal Assoc 11 (1963), pp. 500–541. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (48)

2 L. Dowdney, Annotation: childhood bereavement following parental death, J Child Psychol Psychiat 41 (7) (2000), pp. 819–830. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)

3 K. Kirwin and V. Hamrin, Decreasing the risk of complicated bereavement and future psych disorders in children, J Child Adolesc Psychiat Nurs 18 (2) (2005), pp. 62–78. View Record in Scopus | Cited By in Scopus (10)

4 L. Saler and N. Skoinick, Childhood parental death and depression in adulthood: roles of surviving parent and family environment, Am J Orthopsychiatry 62 (4) (1992), pp. 504–516. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

5 G. Mireault and L. Bond, Parental death in childhood: perceived vulnerability, and adult depression and anxiety, Am J Orthopsychiatry 62 (4) (1992), pp. 517–524. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)

6 G. Christ and A. Christ, Current approaches to helping children cope with a parent's terminal illness, CA Cancer J Clin 56 (2006), pp. 197–212. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

7 P.R. Silverman, Never Too Young to Know: Death in Children's Lives, Oxford University Press, New York (2000).

8 J.W. Warden, Children and Grief: When a Parent Dies, Guilford Press, New York (1996).

9 L. Dowdney, R. Wilson, B. Maughan, M. Allerton, P. Schofield and D. Skuse, Psychological disturbance and service provision in parentally bereaved children: prospective case–control study, BMJ 319 (7206) (1999), pp. 354–357. View Record in Scopus | Cited By in Scopus (25)

10 C. MacPherson, Telling children their ill parent is dying: a study of the factors influencing the well parent, Mortality 10 (2) (2005), pp. 113–120.

11 C. Eaton-Russell, Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers, Temmy Latner Centre for Palliative Care, Toronto (2007).

12 D. Black, Childhood bereavement: distress and long term sequelae can be lessened by early intervention, BMJ 312 (1996), p. 1496. View Record in Scopus | Cited By in Scopus (14)

13 S. Leighton, Bereavement therapy with adolescents—facilitating a process of spiritual growth, J Child Adolesc Psychiatr Nurs 21 (1) (2008), pp. 24–34. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

14 T. Rando, Grief, Dying, and Death: Clinical Interventions for Caregivers, Research Press, Champaign, IL (1984).

15 J. Stokes, Anticipatory grief in families affected by HIV/AIDS, Prog Palliat Care 2 (1994), pp. 43–48.

16 C. Christ and S. Weisenfluh, Parent and child bereavement. In: D. Walsh, Editor, Palliative Medicine (1st ed.), Saunders, Philadelphia (2008).

17 L. Kroll, J. Barnes, A.L. Jones and A. Stein, Cancer in parents: telling children, BMJ 316 (1998), p. 880. View Record in Scopus | Cited By in Scopus (22)

18 J. Piaget, Piaget's theory. In: P. Mussen, Editor, Handbook of Child Psychology (4th ed.), John Wiley & Sons, New York (1970), pp. 703–732.

19 M. Van Riper, Death of a sibling: five sisters, five stories, Pediatr Nurs 23 (6) (1997), pp. 587–593.

20 F. Thompson and S. Payne, Bereaved children's questions to a doctor, Mortality 5 (1) (2000), pp. 74–96. Full Text via CrossRef

21 C.M. Burns, T.W. LeBlanc, A. Abernethy and D. Currow, Young caregivers in the end-of-life setting: a population-based profile of an emerging group, J Palliat Med 13 (10) (2010), pp. 1225–1235. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)

 

 

 
Author and Disclosure Information

S Lawrence Librach, MD, CCFP, FCFP,ab and Heather O'Brien, BA, CLSt Dip, CCLSbc

aTemmy Latner Centre for Palliative Care, Mount Sinai Hospital; bDivision of Palliative Care, University of Toronto; cMax and Beatrice Wolfe Children's Centre, all in Toronto, Canada

Publications
Topics
Legacy Keywords
children's grief; palliative care
Sections
Author and Disclosure Information

S Lawrence Librach, MD, CCFP, FCFP,ab and Heather O'Brien, BA, CLSt Dip, CCLSbc

aTemmy Latner Centre for Palliative Care, Mount Sinai Hospital; bDivision of Palliative Care, University of Toronto; cMax and Beatrice Wolfe Children's Centre, all in Toronto, Canada

Author and Disclosure Information

S Lawrence Librach, MD, CCFP, FCFP,ab and Heather O'Brien, BA, CLSt Dip, CCLSbc

aTemmy Latner Centre for Palliative Care, Mount Sinai Hospital; bDivision of Palliative Care, University of Toronto; cMax and Beatrice Wolfe Children's Centre, all in Toronto, Canada

Children are too often the forgotten mourners in the homes of dying patients. Children, even young children, and youth grieve and mourn the threatened and, then, actual loss of a dying parent, sibling, or other significant family member.1 At a time when the family resources and focus are pulled away and taxed, caregivers are tasked with the difficult job of sorting through their own emotions and a wealth of advice. Caregivers must decide how they will communicate with, include, and support the children/youth in their care.

Although evidence is incomplete and there is a clear need for further studies, links between unresolved childhood grief, or an inability to adequately process their grief, and subsequent psychiatric conditions such as depression and anxiety have been presented as far back as Freud.[2], [3], [4] and [5] In addition, prevalent feelings of responsibility and exclusion and poor communication are consistently identified by researchers interviewing bereaved children/youth about their own experience over the last couple of decades.[6], [7] and [8] Therefore, given the risk of negative psychological and social outcomes associated with children's grief and the struggles communicated by children themselves, it is critical to recognize the important and preventive role of supportive interventions, especially prior to the death of a significant family member.

Looking at caregivers' experiences, there is still a large divide between the advice given by many family and friends in this situation (see Table 1) and what has become accepted within the palliative and grief counseling fields as “best practice.”[6], [7] and [8] In addition, family members' access to professionals trained or knowledgeable in this area is growing but usually still limited.9 Many children/youth are left uninformed, unprepared, and cut off from their family's support.

Table 1. Myths and Realities about Speaking to Children about Grief and Dying. Adapted from MacPherson C.10

 

 

Professionals are not immune to subscribing to the myths listed in Table 1 and “are often inhibited by their anxieties about saying or doing the wrong thing and causing lasting emotional damage.”10 However, by communicating openly and honestly and including children/youth, informed care team members can offer many supportive interventions that a family can benefit from during the time leading up to and following the death. These interventions foster the best outcomes when they are offered early on in the palliative trajectory.[11] and [12]

Our Setting

The Temmy Latner Centre for Palliative Care (TLCPC) at Mount Sinai Hospital in Toronto is one of Canada's largest academic palliative care programs, incorporating a children's center, the Max and Beatrice Wolfe Children's Centre, which provides pediatric palliative care and children's grief programs (Dr. Jay Children's Grief Program). Our children's center supports children, youth, and their families when a family member is dying or has died. This support includes Canada's first Camp Erin, an overnight children's grief camp. Children are referred to these programs for grief counseling by our center's palliative care physicians, local palliative care units and hospices, and a wide variety of community agencies. Children's grief programs are very limited in our large urban setting, as is true in most communities across North America. We have four counselors devoted to child and youth grief support services.

What We Do

Action 1: Intake and Assessment

The center has an open referral policy, accepting referrals from any source, including self-referrals, regardless of the nature of the illness or cause of death.

An intake phone call, lasting between 15 and 60 minutes, is made to the family to assess their needs and to provide psychoeducation and relevant resources. Based on our belief that early intervention provides the most supportive opportunities, families in which the patient is dying are prioritized.

As a result of demand for services being greater than our resources, children and their families bereaved at the time of referral are provided with an initial psychoeducational visit and assessment and then placed on a waiting list if further counseling is deemed appropriate.

In either case, the initial assessment often reveals that what the family needs most is psychoeducation about child/youth grief, communication and development, and reassurance about the benefits of the things they are already doing. Caregivers are provided with educational materials, including a copy of the center's publication Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers,11 a list of Web resources, books, and brochures written by us. All these resources are also available to professionals.

Action 2: Counseling

If further assessment and counseling are warranted, children are seen individually, with siblings and/or with their family depending on the needs and circumstances. The bulk of our counseling services are brief, typically lasting three or four sessions in total. However, more intensive counseling is available on a case-by-case basis, with progress and needs being assessed every four sessions. Counseling techniques including expressive arts, crafts, therapeutic play, and activities are used to support children/youth and families in the grieving process. Families are able to contact counselors when issues arise for them, which often occurs around anniversaries, holidays, other important events, and as children/youth develop and experience their grief in a new light.13

The center's model is resiliency-based, nonpathological, and family-centered. Caregivers are empowered as primary and ongoing sources of support and dominant role models for the grieving child and youth in their care. In addition to conversations with their counselor, families are offered monthly opportunities forcaregiver education and peer support and various therapeutic group activities for children/youth.

Action 3: Complex Cases

Referrals to secondary children's mental health services are facilitated for families with needs beyond the scope of our supportive grief services, including children with indicators of complicated grief who need more intensive counseling. For children with severe psychological distress, referrals are made to a pediatric psychiatrist with special interest in this area.

According to Rando,14 there are a number of forms of unresolved or complicated grief which can overlap, and each has components of denial or regression. These include feelings of grief and mourning being absent, an inhibition of some of the normal symptoms of grief, putting grief on hold for any reason, and when there is a dependent or ambivalent relationship with the deceased. Two common manifestations are extreme anger and extreme guilt.

Some types of death that place children/youth at risk for complicated grief include a sudden or unexpected death, a violent death, a death involving mutilation, the death of a child, and death as the result of a prolonged illness. Also included is complicated loss associated with social stigma such as imprisonment, suicide, AIDS, abortion, severe mental illness, serious family dysfunction, or addiction. However, the presence of these factors does not necessarily lead to complicated grief. Complicated loss is known to be mediated by personal, familial, and social factors that contribute to relative risk and resilience.

What We Say

Engaging a family early in the palliative journey allows greater opportunity to prepare children/youth and prevent possible negative outcomes.[6] and [15] Christ and Weisenfluh16 tell us that the greatest need for support is found during the weeks leading up to the death. A large component of the early intervention we offer is age-appropriate psychoeducation to help caregivers conceptualize how their child/youth may be experiencing and understanding what is happening. Caregivers who anticipate some of the thoughts, feelings, questions, and struggles that their child/youth might face are empowered and children benefit.

The 3 Cs

1. Can I catch it?
2. Did I cause it?
3. Who is going to take care of me?

Julie Stokes15 was able to summarize what children/youth think and worry about most when a family member is dying into three questions. We have coined these three questions the “Three Cs”: Catch, Cause, and Care.

Catch

The first “C” relates to the fear expressed by children/youth that they could catch the illness. Melanie, a 7-year-old, explained that she would have a brain tumor soon because her sister Sarah, who was dying of brain cancer at the time, “is my sister and we lived in the same room always.” If such concerns go unexplored and children are not given clear information, caregivers may see children/youth distance themselves from their ill family member, develop a fear that they and the rest of their family will get sick and die as well, and other implications.17

The weight of language in a child's understanding of illness, death, and dying cannot be emphasized enough.11 More often than not, indirect and generic language used by adults, such as referring to someone as being “sick” or “not doing well,” complicates a child's ability to differentiate between the common cold or flu and life-threatening diseases and illnesses. “My mom was sick and she died; therefore, all people that are sick die.” Many adults believe that their child is too young to understand what cancer means. While it is true that children may not be able to grasp the complex medical information about the illness, they are able to understand a great deal more than they are given credit for, and using the word “cancer” gives them a way to distinguish their dying family member's illness from others such as the common cold or flu.

Cause

The second “C” arises from the common thought in children/youth that they somehow caused or hastened the death and/or prevented the recovery of their family member. Cause is one of the five accepted subconcepts of the developmental understanding of death8 and among the last to be mastered due to its complexity and abstract components. As she sat under her pink bunk-bed, 5-year-old Tayah told her counselor that she had cancer when she was a little girl and then her mother got it after her. Eleven-year-old Joshua shared, “I just have to get to that cancer walk thing. If I can do that, then my mom will get her cure. That is what they said on TV you know, to walk for the cure!”

There are a number of ways that children/youth may try to own responsibility for the illness and death of their family member. They may believe that things they thought or did not think, did or did not do, felt or did not feel were directly related to the cause, progression, or death of the family member.18 These thoughts are especially strong for children in the magical thinking stage of development. In Piaget's theory of cognitive development, magical thinking dominates the preoperational stage (2–7 years old) and describes thinking disconnected from the laws of nature.19 Special attention should be given to assuring children in this age group that they are in no way responsible for the illness or death of their family member, as well as to avoid minimizing these fears and beliefs of responsibility.[6] and [17] This point will need to be readdressed often, with frequent reassurance that they are not responsible.

Care

The third “C” includes concerns about what will become of them and who will take care of them as well as the desire to help care for the dying person. While it is commonly thought that this fear is felt more significantly by children/youth who have a dying or deceased parent, siblings of dying or deceased children appear to be just as challenged. They will experience the same break in their belief that their family member will always be with them.20 Also, many children worry that their surviving parent will be incapacitated and unable to care for them after the death. Four-year-old Alex looked up at his mom one night and said, “You are dying in front of me. I'm scared. Who is going to turn out the light, I can't reach it, and who will cook for me, I can't turn on the stove?” This fear extends beyond who will take over the practical parental roles, to the worry that the child or youth will be orphaned.

Children will often connect their own experience to what they have seen or heard and fear the worst.[18] and [21] Orphans are ever present in children's literature and movies and are often depicted as abandoned to fend for themselves. For many, this fear is grounded in the reality that their current caregiver may not have been emotionally able to address issues of custody or guardianship or that they do not have anyone willing or able to care for the kids after their death. As Alice lay on her bedroom floor writing notes to her dying mother in the critical care unit, she shared, “My mommy and my daddy are dying … they is both going to die,” convinced that this was the truth. Alice's dad is healthy and has been for all of Alice's life.

“Three Cs” and Adolescents

Occasionally, adults have questioned the relativity of the “Three Cs” to our adolescent (ages 13–18) population. Our clinical work has led us to conclude that indeed the “Three Cs” are very real in the lives of the youth we work with. An example of this in relation to “Catch” is Sarah, who expressed distress over the familial traits of breast cancer and the likelihood that her fate will one day mirror that of her mother, aunt, and grandmother.

With regard to “Cause,” we hear youth talk about the relationship between the quality of their caregiving efforts and the death of their family member. Many youth express a feeling that if they had done a better job caregiving, their family member would not have died, especially for youth in primary caregiving roles and single-parent homes.14

When looking at “Care,” youth are impacted by their ability to think abstractly and to experience the loss of what is yet to come. Youth understand that they depend on their caregivers for much more than their practical needs. Joshua talked to his counselor about the loss he was feeling as he searched for employment without his father's help and network. He shared, “If my Dad was here, he would know someone; he would know what I should do.” Important to note is that age is not always the best predictor of cognitive capacity as we see school-aged children grapple with many of the thoughts more commonly expressed by our older population.

Whether or not a caregiver has already spoken with a child about a diagnosis and prognosis, it is helpful to get him or her to consider what the child may be thinking about. Using the foundation of the “Three Cs” will help caregivers understand the importance of open, honest, and concrete communication, as well as feel better equipped to provide their child/youth with support.

Teaching Parents How to Communicate with Their Children

It is difficult for a parent to witness the reaction of a child/youth to such difficult information. This challenge cannot be questioned. However, the benefit to children/youth of having their parents lead or participate in communications and psychoeducation about the cancer and terminal prognosis is clear.[6], [7], [8] and [12] Caregivers are able to clarify assumptions and misperceptions while modeling that it is permissible to talk about cancer and dying together.

Medical staff can facilitate opportunities for such family communications by including children in family meetings and discussions with medical staff and by explaining complex concepts to family members in a simple, concrete fashion. Caregivers often need professionals to welcome and encourage the children to be included. Staff can reassure parents that although they may fear that their children's inclusion will be harmful, on the contrary, inclusion is helpful.

Getting Started

1 Ask what the child/youth understands about the disease/illness.
2 Fill in any gaps in their understanding and explain the treatments that were given.
3 Explain cancer treatments such as chemotherapy, radiation, and surgery.
4 Ask the child/youth what he or she thinks is going to happen.
5 Explain in terms that can be understood that the family member will die.

In any conversation with children/youth about such complex and consequential material, follow their lead in regard to the amount of detail they want. Conceptualize it as an onion: You will want to provide the child/youth with clear and simple language describing basic information. Beyond that, peel off subsequent layers as the child/youth requests more information, always using clear and concrete language. The child/youth may be uncomfortable with the material and may attempt to change the topic. This is a healthy coping mechanism; the average child is quite good at knowing when he or she has reached the limit of their emotional attention span. Allow the child to take the conversation in a different direction, periodically checking to see if he or she is ready to come back to the topic at hand. The benefit of starting this course of conversation early is that it allows children/youth the chance to receive information in small, digestible pieces and the time to process and integrate the information to begin to make meaning of it.

Summary

“What about the kids?” is a dominant and consuming question for caregivers supporting children/youth around the dying and death of a family member. The concerns and fears encompassed in this question can overwhelm caregivers as they put vast amounts of energy into trying to protect children/youth from the suffering and pain that awaits them. Perhaps the hardest lesson these caregivers must learn is that they cannot protect their child/youth from the death any more than they can stop the death from happening. Instead, what is needed most from children/youth is to be included, prepared, and provided with a safe place for emotional expression. Children, as well as adults, will grieve in their own specific way, mediated by their developmental level, circumstances of the illness and death, and protective factors available to them. Providing comprehensive, whole-person care to palliative patients with children/youth in their care ought to include psychoeducation and support for all members of the family. Our communities' bereaved children/youth will be impacted by the death of their family members in countless ways throughout their lives. As professionals caring for people who are dying, we have a responsibility to mediate this impact to the best of our ability.

 

 

References1

1 J. Bowlby, Pathological mourning and childhood mourning, J Am Psychoanal Assoc 11 (1963), pp. 500–541. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (48)

2 L. Dowdney, Annotation: childhood bereavement following parental death, J Child Psychol Psychiat 41 (7) (2000), pp. 819–830. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)

3 K. Kirwin and V. Hamrin, Decreasing the risk of complicated bereavement and future psych disorders in children, J Child Adolesc Psychiat Nurs 18 (2) (2005), pp. 62–78. View Record in Scopus | Cited By in Scopus (10)

4 L. Saler and N. Skoinick, Childhood parental death and depression in adulthood: roles of surviving parent and family environment, Am J Orthopsychiatry 62 (4) (1992), pp. 504–516. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

5 G. Mireault and L. Bond, Parental death in childhood: perceived vulnerability, and adult depression and anxiety, Am J Orthopsychiatry 62 (4) (1992), pp. 517–524. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)

6 G. Christ and A. Christ, Current approaches to helping children cope with a parent's terminal illness, CA Cancer J Clin 56 (2006), pp. 197–212. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

7 P.R. Silverman, Never Too Young to Know: Death in Children's Lives, Oxford University Press, New York (2000).

8 J.W. Warden, Children and Grief: When a Parent Dies, Guilford Press, New York (1996).

9 L. Dowdney, R. Wilson, B. Maughan, M. Allerton, P. Schofield and D. Skuse, Psychological disturbance and service provision in parentally bereaved children: prospective case–control study, BMJ 319 (7206) (1999), pp. 354–357. View Record in Scopus | Cited By in Scopus (25)

10 C. MacPherson, Telling children their ill parent is dying: a study of the factors influencing the well parent, Mortality 10 (2) (2005), pp. 113–120.

11 C. Eaton-Russell, Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers, Temmy Latner Centre for Palliative Care, Toronto (2007).

12 D. Black, Childhood bereavement: distress and long term sequelae can be lessened by early intervention, BMJ 312 (1996), p. 1496. View Record in Scopus | Cited By in Scopus (14)

13 S. Leighton, Bereavement therapy with adolescents—facilitating a process of spiritual growth, J Child Adolesc Psychiatr Nurs 21 (1) (2008), pp. 24–34. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

14 T. Rando, Grief, Dying, and Death: Clinical Interventions for Caregivers, Research Press, Champaign, IL (1984).

15 J. Stokes, Anticipatory grief in families affected by HIV/AIDS, Prog Palliat Care 2 (1994), pp. 43–48.

16 C. Christ and S. Weisenfluh, Parent and child bereavement. In: D. Walsh, Editor, Palliative Medicine (1st ed.), Saunders, Philadelphia (2008).

17 L. Kroll, J. Barnes, A.L. Jones and A. Stein, Cancer in parents: telling children, BMJ 316 (1998), p. 880. View Record in Scopus | Cited By in Scopus (22)

18 J. Piaget, Piaget's theory. In: P. Mussen, Editor, Handbook of Child Psychology (4th ed.), John Wiley & Sons, New York (1970), pp. 703–732.

19 M. Van Riper, Death of a sibling: five sisters, five stories, Pediatr Nurs 23 (6) (1997), pp. 587–593.

20 F. Thompson and S. Payne, Bereaved children's questions to a doctor, Mortality 5 (1) (2000), pp. 74–96. Full Text via CrossRef

21 C.M. Burns, T.W. LeBlanc, A. Abernethy and D. Currow, Young caregivers in the end-of-life setting: a population-based profile of an emerging group, J Palliat Med 13 (10) (2010), pp. 1225–1235. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)

 

 

 

Children are too often the forgotten mourners in the homes of dying patients. Children, even young children, and youth grieve and mourn the threatened and, then, actual loss of a dying parent, sibling, or other significant family member.1 At a time when the family resources and focus are pulled away and taxed, caregivers are tasked with the difficult job of sorting through their own emotions and a wealth of advice. Caregivers must decide how they will communicate with, include, and support the children/youth in their care.

Although evidence is incomplete and there is a clear need for further studies, links between unresolved childhood grief, or an inability to adequately process their grief, and subsequent psychiatric conditions such as depression and anxiety have been presented as far back as Freud.[2], [3], [4] and [5] In addition, prevalent feelings of responsibility and exclusion and poor communication are consistently identified by researchers interviewing bereaved children/youth about their own experience over the last couple of decades.[6], [7] and [8] Therefore, given the risk of negative psychological and social outcomes associated with children's grief and the struggles communicated by children themselves, it is critical to recognize the important and preventive role of supportive interventions, especially prior to the death of a significant family member.

Looking at caregivers' experiences, there is still a large divide between the advice given by many family and friends in this situation (see Table 1) and what has become accepted within the palliative and grief counseling fields as “best practice.”[6], [7] and [8] In addition, family members' access to professionals trained or knowledgeable in this area is growing but usually still limited.9 Many children/youth are left uninformed, unprepared, and cut off from their family's support.

Table 1. Myths and Realities about Speaking to Children about Grief and Dying. Adapted from MacPherson C.10

 

 

Professionals are not immune to subscribing to the myths listed in Table 1 and “are often inhibited by their anxieties about saying or doing the wrong thing and causing lasting emotional damage.”10 However, by communicating openly and honestly and including children/youth, informed care team members can offer many supportive interventions that a family can benefit from during the time leading up to and following the death. These interventions foster the best outcomes when they are offered early on in the palliative trajectory.[11] and [12]

Our Setting

The Temmy Latner Centre for Palliative Care (TLCPC) at Mount Sinai Hospital in Toronto is one of Canada's largest academic palliative care programs, incorporating a children's center, the Max and Beatrice Wolfe Children's Centre, which provides pediatric palliative care and children's grief programs (Dr. Jay Children's Grief Program). Our children's center supports children, youth, and their families when a family member is dying or has died. This support includes Canada's first Camp Erin, an overnight children's grief camp. Children are referred to these programs for grief counseling by our center's palliative care physicians, local palliative care units and hospices, and a wide variety of community agencies. Children's grief programs are very limited in our large urban setting, as is true in most communities across North America. We have four counselors devoted to child and youth grief support services.

What We Do

Action 1: Intake and Assessment

The center has an open referral policy, accepting referrals from any source, including self-referrals, regardless of the nature of the illness or cause of death.

An intake phone call, lasting between 15 and 60 minutes, is made to the family to assess their needs and to provide psychoeducation and relevant resources. Based on our belief that early intervention provides the most supportive opportunities, families in which the patient is dying are prioritized.

As a result of demand for services being greater than our resources, children and their families bereaved at the time of referral are provided with an initial psychoeducational visit and assessment and then placed on a waiting list if further counseling is deemed appropriate.

In either case, the initial assessment often reveals that what the family needs most is psychoeducation about child/youth grief, communication and development, and reassurance about the benefits of the things they are already doing. Caregivers are provided with educational materials, including a copy of the center's publication Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers,11 a list of Web resources, books, and brochures written by us. All these resources are also available to professionals.

Action 2: Counseling

If further assessment and counseling are warranted, children are seen individually, with siblings and/or with their family depending on the needs and circumstances. The bulk of our counseling services are brief, typically lasting three or four sessions in total. However, more intensive counseling is available on a case-by-case basis, with progress and needs being assessed every four sessions. Counseling techniques including expressive arts, crafts, therapeutic play, and activities are used to support children/youth and families in the grieving process. Families are able to contact counselors when issues arise for them, which often occurs around anniversaries, holidays, other important events, and as children/youth develop and experience their grief in a new light.13

The center's model is resiliency-based, nonpathological, and family-centered. Caregivers are empowered as primary and ongoing sources of support and dominant role models for the grieving child and youth in their care. In addition to conversations with their counselor, families are offered monthly opportunities forcaregiver education and peer support and various therapeutic group activities for children/youth.

Action 3: Complex Cases

Referrals to secondary children's mental health services are facilitated for families with needs beyond the scope of our supportive grief services, including children with indicators of complicated grief who need more intensive counseling. For children with severe psychological distress, referrals are made to a pediatric psychiatrist with special interest in this area.

According to Rando,14 there are a number of forms of unresolved or complicated grief which can overlap, and each has components of denial or regression. These include feelings of grief and mourning being absent, an inhibition of some of the normal symptoms of grief, putting grief on hold for any reason, and when there is a dependent or ambivalent relationship with the deceased. Two common manifestations are extreme anger and extreme guilt.

Some types of death that place children/youth at risk for complicated grief include a sudden or unexpected death, a violent death, a death involving mutilation, the death of a child, and death as the result of a prolonged illness. Also included is complicated loss associated with social stigma such as imprisonment, suicide, AIDS, abortion, severe mental illness, serious family dysfunction, or addiction. However, the presence of these factors does not necessarily lead to complicated grief. Complicated loss is known to be mediated by personal, familial, and social factors that contribute to relative risk and resilience.

What We Say

Engaging a family early in the palliative journey allows greater opportunity to prepare children/youth and prevent possible negative outcomes.[6] and [15] Christ and Weisenfluh16 tell us that the greatest need for support is found during the weeks leading up to the death. A large component of the early intervention we offer is age-appropriate psychoeducation to help caregivers conceptualize how their child/youth may be experiencing and understanding what is happening. Caregivers who anticipate some of the thoughts, feelings, questions, and struggles that their child/youth might face are empowered and children benefit.

The 3 Cs

1. Can I catch it?
2. Did I cause it?
3. Who is going to take care of me?

Julie Stokes15 was able to summarize what children/youth think and worry about most when a family member is dying into three questions. We have coined these three questions the “Three Cs”: Catch, Cause, and Care.

Catch

The first “C” relates to the fear expressed by children/youth that they could catch the illness. Melanie, a 7-year-old, explained that she would have a brain tumor soon because her sister Sarah, who was dying of brain cancer at the time, “is my sister and we lived in the same room always.” If such concerns go unexplored and children are not given clear information, caregivers may see children/youth distance themselves from their ill family member, develop a fear that they and the rest of their family will get sick and die as well, and other implications.17

The weight of language in a child's understanding of illness, death, and dying cannot be emphasized enough.11 More often than not, indirect and generic language used by adults, such as referring to someone as being “sick” or “not doing well,” complicates a child's ability to differentiate between the common cold or flu and life-threatening diseases and illnesses. “My mom was sick and she died; therefore, all people that are sick die.” Many adults believe that their child is too young to understand what cancer means. While it is true that children may not be able to grasp the complex medical information about the illness, they are able to understand a great deal more than they are given credit for, and using the word “cancer” gives them a way to distinguish their dying family member's illness from others such as the common cold or flu.

Cause

The second “C” arises from the common thought in children/youth that they somehow caused or hastened the death and/or prevented the recovery of their family member. Cause is one of the five accepted subconcepts of the developmental understanding of death8 and among the last to be mastered due to its complexity and abstract components. As she sat under her pink bunk-bed, 5-year-old Tayah told her counselor that she had cancer when she was a little girl and then her mother got it after her. Eleven-year-old Joshua shared, “I just have to get to that cancer walk thing. If I can do that, then my mom will get her cure. That is what they said on TV you know, to walk for the cure!”

There are a number of ways that children/youth may try to own responsibility for the illness and death of their family member. They may believe that things they thought or did not think, did or did not do, felt or did not feel were directly related to the cause, progression, or death of the family member.18 These thoughts are especially strong for children in the magical thinking stage of development. In Piaget's theory of cognitive development, magical thinking dominates the preoperational stage (2–7 years old) and describes thinking disconnected from the laws of nature.19 Special attention should be given to assuring children in this age group that they are in no way responsible for the illness or death of their family member, as well as to avoid minimizing these fears and beliefs of responsibility.[6] and [17] This point will need to be readdressed often, with frequent reassurance that they are not responsible.

Care

The third “C” includes concerns about what will become of them and who will take care of them as well as the desire to help care for the dying person. While it is commonly thought that this fear is felt more significantly by children/youth who have a dying or deceased parent, siblings of dying or deceased children appear to be just as challenged. They will experience the same break in their belief that their family member will always be with them.20 Also, many children worry that their surviving parent will be incapacitated and unable to care for them after the death. Four-year-old Alex looked up at his mom one night and said, “You are dying in front of me. I'm scared. Who is going to turn out the light, I can't reach it, and who will cook for me, I can't turn on the stove?” This fear extends beyond who will take over the practical parental roles, to the worry that the child or youth will be orphaned.

Children will often connect their own experience to what they have seen or heard and fear the worst.[18] and [21] Orphans are ever present in children's literature and movies and are often depicted as abandoned to fend for themselves. For many, this fear is grounded in the reality that their current caregiver may not have been emotionally able to address issues of custody or guardianship or that they do not have anyone willing or able to care for the kids after their death. As Alice lay on her bedroom floor writing notes to her dying mother in the critical care unit, she shared, “My mommy and my daddy are dying … they is both going to die,” convinced that this was the truth. Alice's dad is healthy and has been for all of Alice's life.

“Three Cs” and Adolescents

Occasionally, adults have questioned the relativity of the “Three Cs” to our adolescent (ages 13–18) population. Our clinical work has led us to conclude that indeed the “Three Cs” are very real in the lives of the youth we work with. An example of this in relation to “Catch” is Sarah, who expressed distress over the familial traits of breast cancer and the likelihood that her fate will one day mirror that of her mother, aunt, and grandmother.

With regard to “Cause,” we hear youth talk about the relationship between the quality of their caregiving efforts and the death of their family member. Many youth express a feeling that if they had done a better job caregiving, their family member would not have died, especially for youth in primary caregiving roles and single-parent homes.14

When looking at “Care,” youth are impacted by their ability to think abstractly and to experience the loss of what is yet to come. Youth understand that they depend on their caregivers for much more than their practical needs. Joshua talked to his counselor about the loss he was feeling as he searched for employment without his father's help and network. He shared, “If my Dad was here, he would know someone; he would know what I should do.” Important to note is that age is not always the best predictor of cognitive capacity as we see school-aged children grapple with many of the thoughts more commonly expressed by our older population.

Whether or not a caregiver has already spoken with a child about a diagnosis and prognosis, it is helpful to get him or her to consider what the child may be thinking about. Using the foundation of the “Three Cs” will help caregivers understand the importance of open, honest, and concrete communication, as well as feel better equipped to provide their child/youth with support.

Teaching Parents How to Communicate with Their Children

It is difficult for a parent to witness the reaction of a child/youth to such difficult information. This challenge cannot be questioned. However, the benefit to children/youth of having their parents lead or participate in communications and psychoeducation about the cancer and terminal prognosis is clear.[6], [7], [8] and [12] Caregivers are able to clarify assumptions and misperceptions while modeling that it is permissible to talk about cancer and dying together.

Medical staff can facilitate opportunities for such family communications by including children in family meetings and discussions with medical staff and by explaining complex concepts to family members in a simple, concrete fashion. Caregivers often need professionals to welcome and encourage the children to be included. Staff can reassure parents that although they may fear that their children's inclusion will be harmful, on the contrary, inclusion is helpful.

Getting Started

1 Ask what the child/youth understands about the disease/illness.
2 Fill in any gaps in their understanding and explain the treatments that were given.
3 Explain cancer treatments such as chemotherapy, radiation, and surgery.
4 Ask the child/youth what he or she thinks is going to happen.
5 Explain in terms that can be understood that the family member will die.

In any conversation with children/youth about such complex and consequential material, follow their lead in regard to the amount of detail they want. Conceptualize it as an onion: You will want to provide the child/youth with clear and simple language describing basic information. Beyond that, peel off subsequent layers as the child/youth requests more information, always using clear and concrete language. The child/youth may be uncomfortable with the material and may attempt to change the topic. This is a healthy coping mechanism; the average child is quite good at knowing when he or she has reached the limit of their emotional attention span. Allow the child to take the conversation in a different direction, periodically checking to see if he or she is ready to come back to the topic at hand. The benefit of starting this course of conversation early is that it allows children/youth the chance to receive information in small, digestible pieces and the time to process and integrate the information to begin to make meaning of it.

Summary

“What about the kids?” is a dominant and consuming question for caregivers supporting children/youth around the dying and death of a family member. The concerns and fears encompassed in this question can overwhelm caregivers as they put vast amounts of energy into trying to protect children/youth from the suffering and pain that awaits them. Perhaps the hardest lesson these caregivers must learn is that they cannot protect their child/youth from the death any more than they can stop the death from happening. Instead, what is needed most from children/youth is to be included, prepared, and provided with a safe place for emotional expression. Children, as well as adults, will grieve in their own specific way, mediated by their developmental level, circumstances of the illness and death, and protective factors available to them. Providing comprehensive, whole-person care to palliative patients with children/youth in their care ought to include psychoeducation and support for all members of the family. Our communities' bereaved children/youth will be impacted by the death of their family members in countless ways throughout their lives. As professionals caring for people who are dying, we have a responsibility to mediate this impact to the best of our ability.

 

 

References1

1 J. Bowlby, Pathological mourning and childhood mourning, J Am Psychoanal Assoc 11 (1963), pp. 500–541. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (48)

2 L. Dowdney, Annotation: childhood bereavement following parental death, J Child Psychol Psychiat 41 (7) (2000), pp. 819–830. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)

3 K. Kirwin and V. Hamrin, Decreasing the risk of complicated bereavement and future psych disorders in children, J Child Adolesc Psychiat Nurs 18 (2) (2005), pp. 62–78. View Record in Scopus | Cited By in Scopus (10)

4 L. Saler and N. Skoinick, Childhood parental death and depression in adulthood: roles of surviving parent and family environment, Am J Orthopsychiatry 62 (4) (1992), pp. 504–516. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

5 G. Mireault and L. Bond, Parental death in childhood: perceived vulnerability, and adult depression and anxiety, Am J Orthopsychiatry 62 (4) (1992), pp. 517–524. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)

6 G. Christ and A. Christ, Current approaches to helping children cope with a parent's terminal illness, CA Cancer J Clin 56 (2006), pp. 197–212. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

7 P.R. Silverman, Never Too Young to Know: Death in Children's Lives, Oxford University Press, New York (2000).

8 J.W. Warden, Children and Grief: When a Parent Dies, Guilford Press, New York (1996).

9 L. Dowdney, R. Wilson, B. Maughan, M. Allerton, P. Schofield and D. Skuse, Psychological disturbance and service provision in parentally bereaved children: prospective case–control study, BMJ 319 (7206) (1999), pp. 354–357. View Record in Scopus | Cited By in Scopus (25)

10 C. MacPherson, Telling children their ill parent is dying: a study of the factors influencing the well parent, Mortality 10 (2) (2005), pp. 113–120.

11 C. Eaton-Russell, Living Dying: A Guide for Adults Supporting Grieving Children and Teenagers, Temmy Latner Centre for Palliative Care, Toronto (2007).

12 D. Black, Childhood bereavement: distress and long term sequelae can be lessened by early intervention, BMJ 312 (1996), p. 1496. View Record in Scopus | Cited By in Scopus (14)

13 S. Leighton, Bereavement therapy with adolescents—facilitating a process of spiritual growth, J Child Adolesc Psychiatr Nurs 21 (1) (2008), pp. 24–34. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

14 T. Rando, Grief, Dying, and Death: Clinical Interventions for Caregivers, Research Press, Champaign, IL (1984).

15 J. Stokes, Anticipatory grief in families affected by HIV/AIDS, Prog Palliat Care 2 (1994), pp. 43–48.

16 C. Christ and S. Weisenfluh, Parent and child bereavement. In: D. Walsh, Editor, Palliative Medicine (1st ed.), Saunders, Philadelphia (2008).

17 L. Kroll, J. Barnes, A.L. Jones and A. Stein, Cancer in parents: telling children, BMJ 316 (1998), p. 880. View Record in Scopus | Cited By in Scopus (22)

18 J. Piaget, Piaget's theory. In: P. Mussen, Editor, Handbook of Child Psychology (4th ed.), John Wiley & Sons, New York (1970), pp. 703–732.

19 M. Van Riper, Death of a sibling: five sisters, five stories, Pediatr Nurs 23 (6) (1997), pp. 587–593.

20 F. Thompson and S. Payne, Bereaved children's questions to a doctor, Mortality 5 (1) (2000), pp. 74–96. Full Text via CrossRef

21 C.M. Burns, T.W. LeBlanc, A. Abernethy and D. Currow, Young caregivers in the end-of-life setting: a population-based profile of an emerging group, J Palliat Med 13 (10) (2010), pp. 1225–1235. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)

 

 

 
Publications
Publications
Topics
Article Type
Display Headline
Supporting Children's Grief within an Adult and Pediatric Palliative Care Program
Display Headline
Supporting Children's Grief within an Adult and Pediatric Palliative Care Program
Legacy Keywords
children's grief; palliative care
Legacy Keywords
children's grief; palliative care
Sections
Citation Override
J Support Oncol. 2011;9(4):136-140
Disallow All Ads
Alternative CME

Providing Pain and Palliative Care Education Internationally

Article Type
Changed
Tue, 05/21/2019 - 12:20
Display Headline
Providing Pain and Palliative Care Education Internationally

 

The Journal of Supportive Oncology
Volume 9, Issue 4, July-August 2011, Pages 129-133

 


 

How we do it

Providing Pain and Palliative Care Education Internationally

Judith A. Paice PhD, RN

, Carma Erickson-Hurt MSN, APRN, ACHPN, Betty Ferrell PhD, MA, FAAN, FPCN, CPCN, Nessa Coyle PhD, ACHPN, FAAN, Patrick J. Coyne MSN, APRN, FAAN, Carol O. Long PhD, RN, FPCN, Polly Mazanec PhD, ACNP, AOCN, FPCN, Pam Malloy RN, MN, OCN, FPCN and Thomas J. Smith MD, FACP

Received 21 December 2010; 
accepted 8 April 2011. 

Available online 2 July 2011.

 

 

Article Outline

Do Your Homework
Health-Care Structure
Available Medications
Education of Health-Care Professionals
Plan the Curriculum and the Program
Personal Considerations
During the Experience
Afterward
Conclusion
Acknowledgements
References
Vitae

 

For many clinicians in oncology, educating other health-care professionals about cancer pain and palliative care is part of their professional life. The need for education exists across clinical settings around the world. Improved education is an urgent need as the prevalence of cancer is increasing. This burden is largely carried by the developing world, where resources are often limited.[1] Global educational efforts, including managing common symptoms, communication, care at the time of death, grief, and other topics, are imperative to reduce pain and suffering.[2] International training efforts require additional expertise and preparation beyond the standard teaching skills needed for all professional education.

The goal of international training efforts in pain and palliative care is to provide useful, culturally relevant programs while empowering participants to sustain these efforts in the long term. Global efforts in palliative care have demonstrated that sharing educational materials, resources, support and encouragement with our international colleagues can provide mentorship to go beyond simply attending a course to developing and expanding their own programs of palliative care in oncology.[3] and [4] To do this well, the following provides specific suggestions for before, during, and after international palliative care training experiences.

Do Your Homework

Before a course, it is essential to learn as much as possible about the region, the culture(s), and the health-care system. Several resources for this information are listed in Table 1. Additionally, speaking with colleagues who have traveled to the country or to those who have emigrated from the country can provide valuable insight. These individuals can provide a wealth of information to assist in developing an appropriate curriculum and specific presentations. As demographics vary, it is important to know the common cancers and other leading causes of death in the region. Issues that may be seen as “competing” issues HIV/AIDS, malaria, immunizations, lack of clean water, or maternal–infant mortality.[5] and [6] Literature, including fiction and nonfiction, as well as movies and other media, can enlighten the traveler regarding life in the region. Local consulates offer opportunities for learning, as do organizations such as the Council on Global Relations. There are rapid changes in global politics, health-care systems, and governments, so it is also vital to have current information.

 

Table 1. Resources for International Educational Efforts
American Society for Clinical Oncology (ASCO)Offers international cancer courses as well as fellowships and other awards.
Centers for Disease Control and Prevention (CDC), http://wwwnc.cdc.gov/travel/Provides information regarding common infectious illnesses, traveler's alerts.
Central Intelligence Agency (CIA), The World Factbook, https://www.cia.gov/library/publications/the-world-factbook/Excellent review of a country's political, demographic, geographic, and other attributes.
City of Hope Pain & Palliative Care Resource Center, http://prc.coh.org/Provides a clearinghouse that includes a wide array of resources and references to enhance pain and palliative care education and research.
End of Life Nursing Education Consortium (ELNEC), http://www.aacn.nche.edu/elnec/Includes relevant articles, resources, and a summary of current international ELNEC training programs.
International Association for Hospice and Palliative Care (IAHPC), http://www.hospicecare.com/Numerous global palliative care resources, including List of Essential Medicines, Global Directory of Educational Programs in Palliative Care, Global Directory of Palliative Care Providers/Services/Organizations, as well as Palliative Care in the Developing World: Principles and Practice.
International Association for the Study of Pain (IASP), http://www.iasp-pain.org/Strong emphasis on support of developing countries with research and educational grants; publishes a Guide to Pain Management in Low-Resource Settings offered without cost.
Open Society Institute–International Palliative Care Initiative, http://www.soros.org/initiatives/health/focus/ipci/aboutOffers support for training, clinical care, and research in palliative care, alone and in collaboration with other organizations.
Pain & Policy Studies Group, http://www.painpolicy.wisc.edu/Excellent resource for information regarding opioid consumption by country as well as guidelines for policies that allow access to necessary medications.
U.S. Department of State, http://www.usembassy.gov/ Bureau of Consular Affairs, http://travel.state.gov/travel/travel_1744.htmlComprehensive lists of US embassies, consulates, and diplomatic missions; information to assist travelers from the United States to other countries, including visa requirements and safety alerts.
World Health Organization, http://www.who.intMany useful resources, including Access to Analgesics and to other Controlled Medicines, as well as statistics regarding common illnesses by country.
 

 


Health-Care Structure

 

Understand the existing health-care structure and what health care is available to all or for select populations. What is the extent of health-care services? Are there clinics for preventive care, or is most care obtained in the hospital? Is home care available with support from nurses and other professionals? Are emergency services available (eg, does the region have ambulances to transport and emergency departments to accept critically ill patients)? Where do patients obtain medications, and do they have to pay out of pocket for these? Do most people die in the hospital or at home? While websites and government sources are valuable, verify this information with clinicians since the clinical reality may be quite different.

Available Medications

To provide useful guidance in symptom management, it is necessary to have a list of available medications used to treat pain, nausea, dyspnea, constipation/diarrhea, wounds, and other symptoms commonly seen in oncology. Your presentation may need to be modified based upon these available drugs (Table 2). Where do patients obtain medications, and do they pay out of pocket for these? There are limitations on availability and access to opioids around the world.[7] Which opioids are available and actually used? What is the process for obtaining a supply of an opioid for a person with cancer? For example, in some countries, physicians can order only one week's worth of medication at a time. In other countries, patients must obtain opioids from the police station rather than a pharmacy. In several settings, only the patient, not family members, can pick up the medication from the dispensing site. And in a few countries, only parenteral opioids are available. It is also helpful to understand issues such as the prevalence of drug trafficking in the region and how this might affect local drug laws. Are traditional medicines, such as herbal therapies, or other techniques commonly used? It is helpful to be aware of these practices and incorporate them into teaching plans where appropriate.

 

 

Table 2. Questions Regarding Available Medications

• Do your presentations reflect the current formulary available in the country?

• Where do patients obtain medications, and do they pay out of pocket for these?

• What opioids are available, in what routes, and what are actually used?

• What is the process for obtaining a supply of an opioid for a person with cancer?

• What is the prevalence of drug trafficking in the region?

• Are traditional medicines, such as herbal therapies, or other techniques commonly used? Are these used in place of, or in addition to, conventional medications?


Education of Health-Care Professionals

International education in palliative care should consider how physicians, nurses, pharmacists, and others are educated. Is the educational system very traditional and formal, with little interaction between students and teachers? Professionals trained in this manner may be less comfortable when faced with role-play, learning through discussion, or other Socratic educational methods. That does not mean that one should exclude these methods when planning the curriculum but, rather, be prepared for silence and possibly even discomfort when first introduced. Seek guidance from local educators as to what methods will be acceptable.

Who is included in the health-care team? Are psychologists available, and are chaplains considered part of health-care services? What is the relationship between physicians, nurses, and other team members? Is collegiality accepted, or is there a hierarchy that limits true teamwork? What is the status of physicians, nurses, and other professionals in the region? In some areas, physicians are highly regarded and financially compensated accordingly. In other parts of the world, physicians have very low social status, respect, and compensation. Within diverse cultures, compensation and acceptance of tips (or bribes) to see a patient or perform an intervention may be accepted practice. Attitudes toward work hours may differ from the Western perspective. In some cultures, socialization and development of personal relationships may be considered more important than other aspects of the workload.[8]

Planning in advance to know the targeted attendees is helpful. It is advisable to inquire if the hosts might consider inviting representatives from the ministry of health, the appropriate drug institutes, other key government officials, as well as medical, nursing, and pharmacy leaders who can become champions for access to pain relief and palliative care. Having multiple disciplines and leaders from health care and government at the same program can foster ongoing communication and understanding. Include chief educators as they can incorporate this content into their respective curricula.

 

Plan the Curriculum and the Program

The importance of cultural issues when developing content cannot be overstated.[9] Factors that might affect pain expression and language or cultural beliefs about death and dying will greatly impact content for teaching. Be aware of local religious and spiritual beliefs impacting pain and palliative care. Consider issues surrounding disclosure of diagnosis and prognosis. Autonomy may not be the prevailing perspective as seen in North America. Ensure that slides are culturally correct and that pictures and illustrations are appropriate. Having the host country leaders review the curriculum in advance is advisable. Avoid cartoons as these may not translate well. Use case examples, but ensure that they represent the types of patients and scenarios seen by the audience. It is also important to avoid being ethnocentric as Western medicine has much to learn from other approaches. It is very helpful to use case studies from the host country. In some settings, trainers will not have access to computers and projectors, limiting the role of PowerPoint slides. Paper presentations or the use of flip-charts may be more accessible.

Consider the need for translation and, if so, which type will be used. Simultaneous interpretation generally requires a sound booth and headphones for participants, and may be more expensive. Consecutive interpretation requires that the instructor present blocks of information, usually a sentence or two, followed by the interpreter providing the content in the appropriate language. This requires speakers to plan much shorter presentations with up to 50% less content being delivered. In either case, trained interpreters can benefit from seeing the slides in advance so they can prepare and clarify prior to the presentation.

When developing an agenda, inquire about the usual times for breaks and meals, as well as time for prayers or other activities. What is considered a “full day” varies around the world, as does the value of adhering rigidly to a schedule. International education generally means that the agenda is fluid; once you are actually in the country and providing the course, other needs may arise. A common mistake is trying to squeeze in too much content. Ask your host to meet prior to the program and, optimally, plan for time before the course to tour health-care facilities. Arrange for a time to meet with key medical, nursing, pharmacy, and governmental leaders who are not scheduled to attend the meeting but might somehow influence curricula and practice. In some settings, local media may be alerted to generate local interest in the topic. Communicate with your host about these opportunities so that arrangements can be made in advance.

For resource-poor countries, consider asking for donations from colleagues before leaving, including books, CDs, and medical supplies. Check local regulations first, particularly if bringing in medications or equipment. If sending books, some countries require high tariff fees to be paid by the receiver when accepting these packages, creating a financial burden for your hosts. Inquire ahead of time if they have to pay to accept these packages. Additionally, in some resource-poor countries, professionals do not have access to personal or work computers and internet café computers often do not have CD drives. Information on jump drives may be more easily accessible.

Finally, visiting educators may want to pack small gifts to give to hosts and others. These should be easily transported and may include items that represent your city or institution. We have also found bringing candy and small toys to be universally appreciated when visiting pediatric settings. A small portable color printer can be used to print photographs of pediatric patients as some of these children have never seen pictures of themselves. You can also print photographs of participants in the training courses.

Personal Considerations

Several months prior to departure, you should contact your traveler's health information resource to identify which vaccinations and what documents are needed to enter the country. To avoid lost time due to illness, ciprofloxacin and antidiarrheal medicines should be obtained before traveling. It is advisable to update your passport. Some countries require you to have sufficient blank pages in your passport to allow entry into their country. An entry fee paid in cash may be required upon arrival. Travelers should consider the political climate of the country and check the U.S. Department of State website (included in Table 1) for alerts or precautions.

Consider appropriate attire when packing. Clothing should reflect respect for the cultural and religious beliefs of the attendees.

During the Experience

It is very useful to meet with interpreters prior to the presentations to clarify any questions. Translation can be quite complicated. For example, a slide that used the term “caring” was interpreted as “romantic love,” and concepts about suffering and death can take on a cultural meaning. Check with interpreters regularly to determine if the speed of delivery is acceptable. Also, translators may have difficulty with this emotional content. In some instances, interpreters have become tearful and required debriefing after palliative care education events. Consider nonverbal communication and personal space. In some cultures, it may not be appropriate to shake hands or to use two hands. Gestures may have very different meanings in other cultures, so avoid these forms of communication. For example, the “OK” sign commonly used in North America, with the tip of the finger touching the tip of the thumb and the other three fingers extended, is considered an obscene gesture in Brazil.

When using teaching strategies other than lecture, respect that some students may not be comfortable at first with nontraditional approaches. Informal teaching strategies that are valued in North America may be viewed as of poor academic quality in other cultures. Debate and discussion, which may make it seem that the student is questioning a teacher's view, may be seen as disrespectful. At times, eliciting personal reflection and experience can engage the audience. For example, when introducing the topic of communication, health-care professionals in the audience can be asked the following questions:

• If you had cancer, would you want to know?

• How about your prognosis?

• Would you want to know that you had a disease that you could die from?

Following these with “What do you tell your patients?” usually engenders excellent discussion.

We have also found that asking participants to do “homework” can be useful, particularly if the students have been quiet or reluctant to communicate during class. Suggested assignments might include listing the five top barriers to cancer pain management in your setting, describing a difficult death or a death that you made better, or related issues. Reticence to speak during class may be due to discomfort with language skills. Some students feel more comfortable sharing ideas in writing, and these assignments have yielded valuable stories that have helped us to understand their experiences and perspectives.

Since the goal of these educational efforts should be sustained, it is helpful to develop a plan for the future with students. Assist them in identifying goals, as well as action items to meet these goals. Allow time for individual meetings between faculty and students to fine-tune these efforts. This ensures that the educational experience will have a greater likelihood of translation into action. To provide practical assistance, if Internet access is available, spend time with small groups to demonstrate literature searches, useful websites, and other information that will foster continuity.

Faculty should meet after each day of training to modify the planned agenda as needed, to optimally meet the needs of the participants. This also provides needed time to debrief about the day's activities and provide support. Particularly when new to international education, the experience may be overwhelming as the status of health-care in developing countries can cause deep personal reflection.

Finally, celebrate. We have found that many students appreciate the opportunity to have some type of closing ceremony to receive certificates and pins, acknowledge their accomplishments, and encourage their future efforts.

Afterward

E-mail, voiceover Internet services, and videoconferencing software have significantly enhanced global communication. Faculty can make themselves available to the trainees after leaving the country using these technologies. Group conversations via e-mail can help solve problems, provide encouragement, and celebrate successes. Connect attendees with international professional organizations to support ongoing educational efforts. It is very useful to identify the leaders or champions and to plan ongoing support to help sustain their commitment. Many countries do not have professional organizations or support networks. These leaders can exist in isolation and suffer great personal sacrifice to lead palliative care efforts in their country.

Conclusion

When educating about pain and palliative care to a worldwide audience, never make assumptions, expect the unexpected, and be flexible. We have found many of these international teaching experiences to be some of the most exhilarating of our professional lives, providing insight to our own practices and creating lasting relationships with colleagues from around the globe. Ultimately, these efforts will improve care for people with cancer.

 

 

Acknowledgments

The authors acknowledge the American Association of Colleges of Nursing and the City of Hope for their ongoing support of the End-of-Life Nursing Education Consortium training activities, as well as the Oncology Nursing Society Foundation and the Open Society Institute for their support of international educational efforts. They also thank Marian Grant for her input.

References [Pub Med ID in Brackets]

1 A.L. Taylor, L.O. Gostin and K.A. Pagonis, Ensuring effective pain treatment: a national and global perspective, JAMA 299 (2008), pp. 89–91 [18167410]. 

2 K. Crane, Palliative care gains ground in developing countries, J Natl Cancer Inst 102 (21) (2010), pp. 1613–1615 [20966432]. 

3 J.A. Paice, B.R. Ferrell, N. Coyle, P. Coyne and M. Callaway, Global efforts to improve palliative care: the International End-of-Life Nursing Education Consortium training programme, J Adv Nurs 61 (2007), pp. 173–180 [18186909].

4 J.A. Paice, B. Ferrell, N. Coyle, P. Coyne and T. Smith, Living and dying in East Africa: implementing the End-of-Life Nursing Education Consortium curriculum in Tanzania, Clin J Oncol Nurs 14 (2010), pp. 161–166 [20350889]. 

5 C. Olweny, C. Sepulveda, A. Merriman, S. Fonn, M. Borok, T. Ngoma, A. Doh and J. Stjernsward, Desirable services and guidelines for the treatment and palliative care of HIV disease patients with cancer in Africa: a World Health Organization consultation, J Palliat Care 19 (2003), pp. 198–205 [14606333]. 

6 C. Sepulveda, V. Habiyatmbete, J. Amandua, M. Borok, E. Kikule, B. Mudanga and B. Solomon, Quality care at the end of life in Africa, BMJ 327 (2003), pp. 209–213 [12881267]. 

7 E.L. Krakauer, R. Wenk, R. Buitrago, P. Jenkins and W. Scholten, Opioid inaccessibility and its human consequences: reports from the field, J Pain Palliat Care Pharmacother 24 (2010), pp. 239–243 [20718644].

8 C.M. Bolin, Developing a postbasic gerontology program for international learners: considerations for the process, J Contin Educ Nurs 34 (2003), pp. 177–183 [12887229].

9 K.D. Meneses and C.H. Yarbro, Cultural perspectives of international breast health and breast cancer education, J Nurs Scholarsh 39 (2) (2007), pp. 105–112 [19058079]. 

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and none were reported.


Correspondence to: Judith A. Paice, PhD, RN, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair Street, Suite 850, Chicago, IL 60611; telephone: (312) 695-4157; fax: (312) 695-6189.

 


Vitae

Dr. Paice is Director of the Cancer Pain Program, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.

Carma Erickson-Hurt is a faculty member at Grand Canyon University, Phoenix, Arizona.

Dr. Ferrell is a Professor and Research Scientist at the City of Hope National Medical Center, Duarte, California.

Nessa Coyle is on the Pain and Palliative Care Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York.

Dr. Coyne is Clinical Director of the Thomas Palliative Care Program, Virginia Commonwealth University/Massey Cancer Center, Richmond, Virginia.

Dr. Long is a geriatric nursing consultant and codirector of the Palliative Care for Advanced Dementia, Beatitudes Campus, Phoenix, Arizona.

Dr. Mazanec is a clinical nurse specialist at the University Hospitals Seidman Cancer Center, Cleveland, Ohio.

Pam Malloy is ELNEC Project Director, American Association of Colleges of Nursing, Washington, DC.

Dr. Smith is Professor of Medicine and Palliative Care Research, Virginia Commonwealth University/Massey Cancer Center, Richmond.

Author and Disclosure Information

 

 

Publications
Legacy Keywords
Pain, Palliative Care, Cancer, International, Education
Sections
Author and Disclosure Information

 

 

Author and Disclosure Information

 

 

 

The Journal of Supportive Oncology
Volume 9, Issue 4, July-August 2011, Pages 129-133

 


 

How we do it

Providing Pain and Palliative Care Education Internationally

Judith A. Paice PhD, RN

, Carma Erickson-Hurt MSN, APRN, ACHPN, Betty Ferrell PhD, MA, FAAN, FPCN, CPCN, Nessa Coyle PhD, ACHPN, FAAN, Patrick J. Coyne MSN, APRN, FAAN, Carol O. Long PhD, RN, FPCN, Polly Mazanec PhD, ACNP, AOCN, FPCN, Pam Malloy RN, MN, OCN, FPCN and Thomas J. Smith MD, FACP

Received 21 December 2010; 
accepted 8 April 2011. 

Available online 2 July 2011.

 

 

Article Outline

Do Your Homework
Health-Care Structure
Available Medications
Education of Health-Care Professionals
Plan the Curriculum and the Program
Personal Considerations
During the Experience
Afterward
Conclusion
Acknowledgements
References
Vitae

 

For many clinicians in oncology, educating other health-care professionals about cancer pain and palliative care is part of their professional life. The need for education exists across clinical settings around the world. Improved education is an urgent need as the prevalence of cancer is increasing. This burden is largely carried by the developing world, where resources are often limited.[1] Global educational efforts, including managing common symptoms, communication, care at the time of death, grief, and other topics, are imperative to reduce pain and suffering.[2] International training efforts require additional expertise and preparation beyond the standard teaching skills needed for all professional education.

The goal of international training efforts in pain and palliative care is to provide useful, culturally relevant programs while empowering participants to sustain these efforts in the long term. Global efforts in palliative care have demonstrated that sharing educational materials, resources, support and encouragement with our international colleagues can provide mentorship to go beyond simply attending a course to developing and expanding their own programs of palliative care in oncology.[3] and [4] To do this well, the following provides specific suggestions for before, during, and after international palliative care training experiences.

Do Your Homework

Before a course, it is essential to learn as much as possible about the region, the culture(s), and the health-care system. Several resources for this information are listed in Table 1. Additionally, speaking with colleagues who have traveled to the country or to those who have emigrated from the country can provide valuable insight. These individuals can provide a wealth of information to assist in developing an appropriate curriculum and specific presentations. As demographics vary, it is important to know the common cancers and other leading causes of death in the region. Issues that may be seen as “competing” issues HIV/AIDS, malaria, immunizations, lack of clean water, or maternal–infant mortality.[5] and [6] Literature, including fiction and nonfiction, as well as movies and other media, can enlighten the traveler regarding life in the region. Local consulates offer opportunities for learning, as do organizations such as the Council on Global Relations. There are rapid changes in global politics, health-care systems, and governments, so it is also vital to have current information.

 

Table 1. Resources for International Educational Efforts
American Society for Clinical Oncology (ASCO)Offers international cancer courses as well as fellowships and other awards.
Centers for Disease Control and Prevention (CDC), http://wwwnc.cdc.gov/travel/Provides information regarding common infectious illnesses, traveler's alerts.
Central Intelligence Agency (CIA), The World Factbook, https://www.cia.gov/library/publications/the-world-factbook/Excellent review of a country's political, demographic, geographic, and other attributes.
City of Hope Pain & Palliative Care Resource Center, http://prc.coh.org/Provides a clearinghouse that includes a wide array of resources and references to enhance pain and palliative care education and research.
End of Life Nursing Education Consortium (ELNEC), http://www.aacn.nche.edu/elnec/Includes relevant articles, resources, and a summary of current international ELNEC training programs.
International Association for Hospice and Palliative Care (IAHPC), http://www.hospicecare.com/Numerous global palliative care resources, including List of Essential Medicines, Global Directory of Educational Programs in Palliative Care, Global Directory of Palliative Care Providers/Services/Organizations, as well as Palliative Care in the Developing World: Principles and Practice.
International Association for the Study of Pain (IASP), http://www.iasp-pain.org/Strong emphasis on support of developing countries with research and educational grants; publishes a Guide to Pain Management in Low-Resource Settings offered without cost.
Open Society Institute–International Palliative Care Initiative, http://www.soros.org/initiatives/health/focus/ipci/aboutOffers support for training, clinical care, and research in palliative care, alone and in collaboration with other organizations.
Pain & Policy Studies Group, http://www.painpolicy.wisc.edu/Excellent resource for information regarding opioid consumption by country as well as guidelines for policies that allow access to necessary medications.
U.S. Department of State, http://www.usembassy.gov/ Bureau of Consular Affairs, http://travel.state.gov/travel/travel_1744.htmlComprehensive lists of US embassies, consulates, and diplomatic missions; information to assist travelers from the United States to other countries, including visa requirements and safety alerts.
World Health Organization, http://www.who.intMany useful resources, including Access to Analgesics and to other Controlled Medicines, as well as statistics regarding common illnesses by country.
 

 


Health-Care Structure

 

Understand the existing health-care structure and what health care is available to all or for select populations. What is the extent of health-care services? Are there clinics for preventive care, or is most care obtained in the hospital? Is home care available with support from nurses and other professionals? Are emergency services available (eg, does the region have ambulances to transport and emergency departments to accept critically ill patients)? Where do patients obtain medications, and do they have to pay out of pocket for these? Do most people die in the hospital or at home? While websites and government sources are valuable, verify this information with clinicians since the clinical reality may be quite different.

Available Medications

To provide useful guidance in symptom management, it is necessary to have a list of available medications used to treat pain, nausea, dyspnea, constipation/diarrhea, wounds, and other symptoms commonly seen in oncology. Your presentation may need to be modified based upon these available drugs (Table 2). Where do patients obtain medications, and do they pay out of pocket for these? There are limitations on availability and access to opioids around the world.[7] Which opioids are available and actually used? What is the process for obtaining a supply of an opioid for a person with cancer? For example, in some countries, physicians can order only one week's worth of medication at a time. In other countries, patients must obtain opioids from the police station rather than a pharmacy. In several settings, only the patient, not family members, can pick up the medication from the dispensing site. And in a few countries, only parenteral opioids are available. It is also helpful to understand issues such as the prevalence of drug trafficking in the region and how this might affect local drug laws. Are traditional medicines, such as herbal therapies, or other techniques commonly used? It is helpful to be aware of these practices and incorporate them into teaching plans where appropriate.

 

 

Table 2. Questions Regarding Available Medications

• Do your presentations reflect the current formulary available in the country?

• Where do patients obtain medications, and do they pay out of pocket for these?

• What opioids are available, in what routes, and what are actually used?

• What is the process for obtaining a supply of an opioid for a person with cancer?

• What is the prevalence of drug trafficking in the region?

• Are traditional medicines, such as herbal therapies, or other techniques commonly used? Are these used in place of, or in addition to, conventional medications?


Education of Health-Care Professionals

International education in palliative care should consider how physicians, nurses, pharmacists, and others are educated. Is the educational system very traditional and formal, with little interaction between students and teachers? Professionals trained in this manner may be less comfortable when faced with role-play, learning through discussion, or other Socratic educational methods. That does not mean that one should exclude these methods when planning the curriculum but, rather, be prepared for silence and possibly even discomfort when first introduced. Seek guidance from local educators as to what methods will be acceptable.

Who is included in the health-care team? Are psychologists available, and are chaplains considered part of health-care services? What is the relationship between physicians, nurses, and other team members? Is collegiality accepted, or is there a hierarchy that limits true teamwork? What is the status of physicians, nurses, and other professionals in the region? In some areas, physicians are highly regarded and financially compensated accordingly. In other parts of the world, physicians have very low social status, respect, and compensation. Within diverse cultures, compensation and acceptance of tips (or bribes) to see a patient or perform an intervention may be accepted practice. Attitudes toward work hours may differ from the Western perspective. In some cultures, socialization and development of personal relationships may be considered more important than other aspects of the workload.[8]

Planning in advance to know the targeted attendees is helpful. It is advisable to inquire if the hosts might consider inviting representatives from the ministry of health, the appropriate drug institutes, other key government officials, as well as medical, nursing, and pharmacy leaders who can become champions for access to pain relief and palliative care. Having multiple disciplines and leaders from health care and government at the same program can foster ongoing communication and understanding. Include chief educators as they can incorporate this content into their respective curricula.

 

Plan the Curriculum and the Program

The importance of cultural issues when developing content cannot be overstated.[9] Factors that might affect pain expression and language or cultural beliefs about death and dying will greatly impact content for teaching. Be aware of local religious and spiritual beliefs impacting pain and palliative care. Consider issues surrounding disclosure of diagnosis and prognosis. Autonomy may not be the prevailing perspective as seen in North America. Ensure that slides are culturally correct and that pictures and illustrations are appropriate. Having the host country leaders review the curriculum in advance is advisable. Avoid cartoons as these may not translate well. Use case examples, but ensure that they represent the types of patients and scenarios seen by the audience. It is also important to avoid being ethnocentric as Western medicine has much to learn from other approaches. It is very helpful to use case studies from the host country. In some settings, trainers will not have access to computers and projectors, limiting the role of PowerPoint slides. Paper presentations or the use of flip-charts may be more accessible.

Consider the need for translation and, if so, which type will be used. Simultaneous interpretation generally requires a sound booth and headphones for participants, and may be more expensive. Consecutive interpretation requires that the instructor present blocks of information, usually a sentence or two, followed by the interpreter providing the content in the appropriate language. This requires speakers to plan much shorter presentations with up to 50% less content being delivered. In either case, trained interpreters can benefit from seeing the slides in advance so they can prepare and clarify prior to the presentation.

When developing an agenda, inquire about the usual times for breaks and meals, as well as time for prayers or other activities. What is considered a “full day” varies around the world, as does the value of adhering rigidly to a schedule. International education generally means that the agenda is fluid; once you are actually in the country and providing the course, other needs may arise. A common mistake is trying to squeeze in too much content. Ask your host to meet prior to the program and, optimally, plan for time before the course to tour health-care facilities. Arrange for a time to meet with key medical, nursing, pharmacy, and governmental leaders who are not scheduled to attend the meeting but might somehow influence curricula and practice. In some settings, local media may be alerted to generate local interest in the topic. Communicate with your host about these opportunities so that arrangements can be made in advance.

For resource-poor countries, consider asking for donations from colleagues before leaving, including books, CDs, and medical supplies. Check local regulations first, particularly if bringing in medications or equipment. If sending books, some countries require high tariff fees to be paid by the receiver when accepting these packages, creating a financial burden for your hosts. Inquire ahead of time if they have to pay to accept these packages. Additionally, in some resource-poor countries, professionals do not have access to personal or work computers and internet café computers often do not have CD drives. Information on jump drives may be more easily accessible.

Finally, visiting educators may want to pack small gifts to give to hosts and others. These should be easily transported and may include items that represent your city or institution. We have also found bringing candy and small toys to be universally appreciated when visiting pediatric settings. A small portable color printer can be used to print photographs of pediatric patients as some of these children have never seen pictures of themselves. You can also print photographs of participants in the training courses.

Personal Considerations

Several months prior to departure, you should contact your traveler's health information resource to identify which vaccinations and what documents are needed to enter the country. To avoid lost time due to illness, ciprofloxacin and antidiarrheal medicines should be obtained before traveling. It is advisable to update your passport. Some countries require you to have sufficient blank pages in your passport to allow entry into their country. An entry fee paid in cash may be required upon arrival. Travelers should consider the political climate of the country and check the U.S. Department of State website (included in Table 1) for alerts or precautions.

Consider appropriate attire when packing. Clothing should reflect respect for the cultural and religious beliefs of the attendees.

During the Experience

It is very useful to meet with interpreters prior to the presentations to clarify any questions. Translation can be quite complicated. For example, a slide that used the term “caring” was interpreted as “romantic love,” and concepts about suffering and death can take on a cultural meaning. Check with interpreters regularly to determine if the speed of delivery is acceptable. Also, translators may have difficulty with this emotional content. In some instances, interpreters have become tearful and required debriefing after palliative care education events. Consider nonverbal communication and personal space. In some cultures, it may not be appropriate to shake hands or to use two hands. Gestures may have very different meanings in other cultures, so avoid these forms of communication. For example, the “OK” sign commonly used in North America, with the tip of the finger touching the tip of the thumb and the other three fingers extended, is considered an obscene gesture in Brazil.

When using teaching strategies other than lecture, respect that some students may not be comfortable at first with nontraditional approaches. Informal teaching strategies that are valued in North America may be viewed as of poor academic quality in other cultures. Debate and discussion, which may make it seem that the student is questioning a teacher's view, may be seen as disrespectful. At times, eliciting personal reflection and experience can engage the audience. For example, when introducing the topic of communication, health-care professionals in the audience can be asked the following questions:

• If you had cancer, would you want to know?

• How about your prognosis?

• Would you want to know that you had a disease that you could die from?

Following these with “What do you tell your patients?” usually engenders excellent discussion.

We have also found that asking participants to do “homework” can be useful, particularly if the students have been quiet or reluctant to communicate during class. Suggested assignments might include listing the five top barriers to cancer pain management in your setting, describing a difficult death or a death that you made better, or related issues. Reticence to speak during class may be due to discomfort with language skills. Some students feel more comfortable sharing ideas in writing, and these assignments have yielded valuable stories that have helped us to understand their experiences and perspectives.

Since the goal of these educational efforts should be sustained, it is helpful to develop a plan for the future with students. Assist them in identifying goals, as well as action items to meet these goals. Allow time for individual meetings between faculty and students to fine-tune these efforts. This ensures that the educational experience will have a greater likelihood of translation into action. To provide practical assistance, if Internet access is available, spend time with small groups to demonstrate literature searches, useful websites, and other information that will foster continuity.

Faculty should meet after each day of training to modify the planned agenda as needed, to optimally meet the needs of the participants. This also provides needed time to debrief about the day's activities and provide support. Particularly when new to international education, the experience may be overwhelming as the status of health-care in developing countries can cause deep personal reflection.

Finally, celebrate. We have found that many students appreciate the opportunity to have some type of closing ceremony to receive certificates and pins, acknowledge their accomplishments, and encourage their future efforts.

Afterward

E-mail, voiceover Internet services, and videoconferencing software have significantly enhanced global communication. Faculty can make themselves available to the trainees after leaving the country using these technologies. Group conversations via e-mail can help solve problems, provide encouragement, and celebrate successes. Connect attendees with international professional organizations to support ongoing educational efforts. It is very useful to identify the leaders or champions and to plan ongoing support to help sustain their commitment. Many countries do not have professional organizations or support networks. These leaders can exist in isolation and suffer great personal sacrifice to lead palliative care efforts in their country.

Conclusion

When educating about pain and palliative care to a worldwide audience, never make assumptions, expect the unexpected, and be flexible. We have found many of these international teaching experiences to be some of the most exhilarating of our professional lives, providing insight to our own practices and creating lasting relationships with colleagues from around the globe. Ultimately, these efforts will improve care for people with cancer.

 

 

Acknowledgments

The authors acknowledge the American Association of Colleges of Nursing and the City of Hope for their ongoing support of the End-of-Life Nursing Education Consortium training activities, as well as the Oncology Nursing Society Foundation and the Open Society Institute for their support of international educational efforts. They also thank Marian Grant for her input.

References [Pub Med ID in Brackets]

1 A.L. Taylor, L.O. Gostin and K.A. Pagonis, Ensuring effective pain treatment: a national and global perspective, JAMA 299 (2008), pp. 89–91 [18167410]. 

2 K. Crane, Palliative care gains ground in developing countries, J Natl Cancer Inst 102 (21) (2010), pp. 1613–1615 [20966432]. 

3 J.A. Paice, B.R. Ferrell, N. Coyle, P. Coyne and M. Callaway, Global efforts to improve palliative care: the International End-of-Life Nursing Education Consortium training programme, J Adv Nurs 61 (2007), pp. 173–180 [18186909].

4 J.A. Paice, B. Ferrell, N. Coyle, P. Coyne and T. Smith, Living and dying in East Africa: implementing the End-of-Life Nursing Education Consortium curriculum in Tanzania, Clin J Oncol Nurs 14 (2010), pp. 161–166 [20350889]. 

5 C. Olweny, C. Sepulveda, A. Merriman, S. Fonn, M. Borok, T. Ngoma, A. Doh and J. Stjernsward, Desirable services and guidelines for the treatment and palliative care of HIV disease patients with cancer in Africa: a World Health Organization consultation, J Palliat Care 19 (2003), pp. 198–205 [14606333]. 

6 C. Sepulveda, V. Habiyatmbete, J. Amandua, M. Borok, E. Kikule, B. Mudanga and B. Solomon, Quality care at the end of life in Africa, BMJ 327 (2003), pp. 209–213 [12881267]. 

7 E.L. Krakauer, R. Wenk, R. Buitrago, P. Jenkins and W. Scholten, Opioid inaccessibility and its human consequences: reports from the field, J Pain Palliat Care Pharmacother 24 (2010), pp. 239–243 [20718644].

8 C.M. Bolin, Developing a postbasic gerontology program for international learners: considerations for the process, J Contin Educ Nurs 34 (2003), pp. 177–183 [12887229].

9 K.D. Meneses and C.H. Yarbro, Cultural perspectives of international breast health and breast cancer education, J Nurs Scholarsh 39 (2) (2007), pp. 105–112 [19058079]. 

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and none were reported.


Correspondence to: Judith A. Paice, PhD, RN, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair Street, Suite 850, Chicago, IL 60611; telephone: (312) 695-4157; fax: (312) 695-6189.

 


Vitae

Dr. Paice is Director of the Cancer Pain Program, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.

Carma Erickson-Hurt is a faculty member at Grand Canyon University, Phoenix, Arizona.

Dr. Ferrell is a Professor and Research Scientist at the City of Hope National Medical Center, Duarte, California.

Nessa Coyle is on the Pain and Palliative Care Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York.

Dr. Coyne is Clinical Director of the Thomas Palliative Care Program, Virginia Commonwealth University/Massey Cancer Center, Richmond, Virginia.

Dr. Long is a geriatric nursing consultant and codirector of the Palliative Care for Advanced Dementia, Beatitudes Campus, Phoenix, Arizona.

Dr. Mazanec is a clinical nurse specialist at the University Hospitals Seidman Cancer Center, Cleveland, Ohio.

Pam Malloy is ELNEC Project Director, American Association of Colleges of Nursing, Washington, DC.

Dr. Smith is Professor of Medicine and Palliative Care Research, Virginia Commonwealth University/Massey Cancer Center, Richmond.

 

The Journal of Supportive Oncology
Volume 9, Issue 4, July-August 2011, Pages 129-133

 


 

How we do it

Providing Pain and Palliative Care Education Internationally

Judith A. Paice PhD, RN

, Carma Erickson-Hurt MSN, APRN, ACHPN, Betty Ferrell PhD, MA, FAAN, FPCN, CPCN, Nessa Coyle PhD, ACHPN, FAAN, Patrick J. Coyne MSN, APRN, FAAN, Carol O. Long PhD, RN, FPCN, Polly Mazanec PhD, ACNP, AOCN, FPCN, Pam Malloy RN, MN, OCN, FPCN and Thomas J. Smith MD, FACP

Received 21 December 2010; 
accepted 8 April 2011. 

Available online 2 July 2011.

 

 

Article Outline

Do Your Homework
Health-Care Structure
Available Medications
Education of Health-Care Professionals
Plan the Curriculum and the Program
Personal Considerations
During the Experience
Afterward
Conclusion
Acknowledgements
References
Vitae

 

For many clinicians in oncology, educating other health-care professionals about cancer pain and palliative care is part of their professional life. The need for education exists across clinical settings around the world. Improved education is an urgent need as the prevalence of cancer is increasing. This burden is largely carried by the developing world, where resources are often limited.[1] Global educational efforts, including managing common symptoms, communication, care at the time of death, grief, and other topics, are imperative to reduce pain and suffering.[2] International training efforts require additional expertise and preparation beyond the standard teaching skills needed for all professional education.

The goal of international training efforts in pain and palliative care is to provide useful, culturally relevant programs while empowering participants to sustain these efforts in the long term. Global efforts in palliative care have demonstrated that sharing educational materials, resources, support and encouragement with our international colleagues can provide mentorship to go beyond simply attending a course to developing and expanding their own programs of palliative care in oncology.[3] and [4] To do this well, the following provides specific suggestions for before, during, and after international palliative care training experiences.

Do Your Homework

Before a course, it is essential to learn as much as possible about the region, the culture(s), and the health-care system. Several resources for this information are listed in Table 1. Additionally, speaking with colleagues who have traveled to the country or to those who have emigrated from the country can provide valuable insight. These individuals can provide a wealth of information to assist in developing an appropriate curriculum and specific presentations. As demographics vary, it is important to know the common cancers and other leading causes of death in the region. Issues that may be seen as “competing” issues HIV/AIDS, malaria, immunizations, lack of clean water, or maternal–infant mortality.[5] and [6] Literature, including fiction and nonfiction, as well as movies and other media, can enlighten the traveler regarding life in the region. Local consulates offer opportunities for learning, as do organizations such as the Council on Global Relations. There are rapid changes in global politics, health-care systems, and governments, so it is also vital to have current information.

 

Table 1. Resources for International Educational Efforts
American Society for Clinical Oncology (ASCO)Offers international cancer courses as well as fellowships and other awards.
Centers for Disease Control and Prevention (CDC), http://wwwnc.cdc.gov/travel/Provides information regarding common infectious illnesses, traveler's alerts.
Central Intelligence Agency (CIA), The World Factbook, https://www.cia.gov/library/publications/the-world-factbook/Excellent review of a country's political, demographic, geographic, and other attributes.
City of Hope Pain & Palliative Care Resource Center, http://prc.coh.org/Provides a clearinghouse that includes a wide array of resources and references to enhance pain and palliative care education and research.
End of Life Nursing Education Consortium (ELNEC), http://www.aacn.nche.edu/elnec/Includes relevant articles, resources, and a summary of current international ELNEC training programs.
International Association for Hospice and Palliative Care (IAHPC), http://www.hospicecare.com/Numerous global palliative care resources, including List of Essential Medicines, Global Directory of Educational Programs in Palliative Care, Global Directory of Palliative Care Providers/Services/Organizations, as well as Palliative Care in the Developing World: Principles and Practice.
International Association for the Study of Pain (IASP), http://www.iasp-pain.org/Strong emphasis on support of developing countries with research and educational grants; publishes a Guide to Pain Management in Low-Resource Settings offered without cost.
Open Society Institute–International Palliative Care Initiative, http://www.soros.org/initiatives/health/focus/ipci/aboutOffers support for training, clinical care, and research in palliative care, alone and in collaboration with other organizations.
Pain & Policy Studies Group, http://www.painpolicy.wisc.edu/Excellent resource for information regarding opioid consumption by country as well as guidelines for policies that allow access to necessary medications.
U.S. Department of State, http://www.usembassy.gov/ Bureau of Consular Affairs, http://travel.state.gov/travel/travel_1744.htmlComprehensive lists of US embassies, consulates, and diplomatic missions; information to assist travelers from the United States to other countries, including visa requirements and safety alerts.
World Health Organization, http://www.who.intMany useful resources, including Access to Analgesics and to other Controlled Medicines, as well as statistics regarding common illnesses by country.
 

 


Health-Care Structure

 

Understand the existing health-care structure and what health care is available to all or for select populations. What is the extent of health-care services? Are there clinics for preventive care, or is most care obtained in the hospital? Is home care available with support from nurses and other professionals? Are emergency services available (eg, does the region have ambulances to transport and emergency departments to accept critically ill patients)? Where do patients obtain medications, and do they have to pay out of pocket for these? Do most people die in the hospital or at home? While websites and government sources are valuable, verify this information with clinicians since the clinical reality may be quite different.

Available Medications

To provide useful guidance in symptom management, it is necessary to have a list of available medications used to treat pain, nausea, dyspnea, constipation/diarrhea, wounds, and other symptoms commonly seen in oncology. Your presentation may need to be modified based upon these available drugs (Table 2). Where do patients obtain medications, and do they pay out of pocket for these? There are limitations on availability and access to opioids around the world.[7] Which opioids are available and actually used? What is the process for obtaining a supply of an opioid for a person with cancer? For example, in some countries, physicians can order only one week's worth of medication at a time. In other countries, patients must obtain opioids from the police station rather than a pharmacy. In several settings, only the patient, not family members, can pick up the medication from the dispensing site. And in a few countries, only parenteral opioids are available. It is also helpful to understand issues such as the prevalence of drug trafficking in the region and how this might affect local drug laws. Are traditional medicines, such as herbal therapies, or other techniques commonly used? It is helpful to be aware of these practices and incorporate them into teaching plans where appropriate.

 

 

Table 2. Questions Regarding Available Medications

• Do your presentations reflect the current formulary available in the country?

• Where do patients obtain medications, and do they pay out of pocket for these?

• What opioids are available, in what routes, and what are actually used?

• What is the process for obtaining a supply of an opioid for a person with cancer?

• What is the prevalence of drug trafficking in the region?

• Are traditional medicines, such as herbal therapies, or other techniques commonly used? Are these used in place of, or in addition to, conventional medications?


Education of Health-Care Professionals

International education in palliative care should consider how physicians, nurses, pharmacists, and others are educated. Is the educational system very traditional and formal, with little interaction between students and teachers? Professionals trained in this manner may be less comfortable when faced with role-play, learning through discussion, or other Socratic educational methods. That does not mean that one should exclude these methods when planning the curriculum but, rather, be prepared for silence and possibly even discomfort when first introduced. Seek guidance from local educators as to what methods will be acceptable.

Who is included in the health-care team? Are psychologists available, and are chaplains considered part of health-care services? What is the relationship between physicians, nurses, and other team members? Is collegiality accepted, or is there a hierarchy that limits true teamwork? What is the status of physicians, nurses, and other professionals in the region? In some areas, physicians are highly regarded and financially compensated accordingly. In other parts of the world, physicians have very low social status, respect, and compensation. Within diverse cultures, compensation and acceptance of tips (or bribes) to see a patient or perform an intervention may be accepted practice. Attitudes toward work hours may differ from the Western perspective. In some cultures, socialization and development of personal relationships may be considered more important than other aspects of the workload.[8]

Planning in advance to know the targeted attendees is helpful. It is advisable to inquire if the hosts might consider inviting representatives from the ministry of health, the appropriate drug institutes, other key government officials, as well as medical, nursing, and pharmacy leaders who can become champions for access to pain relief and palliative care. Having multiple disciplines and leaders from health care and government at the same program can foster ongoing communication and understanding. Include chief educators as they can incorporate this content into their respective curricula.

 

Plan the Curriculum and the Program

The importance of cultural issues when developing content cannot be overstated.[9] Factors that might affect pain expression and language or cultural beliefs about death and dying will greatly impact content for teaching. Be aware of local religious and spiritual beliefs impacting pain and palliative care. Consider issues surrounding disclosure of diagnosis and prognosis. Autonomy may not be the prevailing perspective as seen in North America. Ensure that slides are culturally correct and that pictures and illustrations are appropriate. Having the host country leaders review the curriculum in advance is advisable. Avoid cartoons as these may not translate well. Use case examples, but ensure that they represent the types of patients and scenarios seen by the audience. It is also important to avoid being ethnocentric as Western medicine has much to learn from other approaches. It is very helpful to use case studies from the host country. In some settings, trainers will not have access to computers and projectors, limiting the role of PowerPoint slides. Paper presentations or the use of flip-charts may be more accessible.

Consider the need for translation and, if so, which type will be used. Simultaneous interpretation generally requires a sound booth and headphones for participants, and may be more expensive. Consecutive interpretation requires that the instructor present blocks of information, usually a sentence or two, followed by the interpreter providing the content in the appropriate language. This requires speakers to plan much shorter presentations with up to 50% less content being delivered. In either case, trained interpreters can benefit from seeing the slides in advance so they can prepare and clarify prior to the presentation.

When developing an agenda, inquire about the usual times for breaks and meals, as well as time for prayers or other activities. What is considered a “full day” varies around the world, as does the value of adhering rigidly to a schedule. International education generally means that the agenda is fluid; once you are actually in the country and providing the course, other needs may arise. A common mistake is trying to squeeze in too much content. Ask your host to meet prior to the program and, optimally, plan for time before the course to tour health-care facilities. Arrange for a time to meet with key medical, nursing, pharmacy, and governmental leaders who are not scheduled to attend the meeting but might somehow influence curricula and practice. In some settings, local media may be alerted to generate local interest in the topic. Communicate with your host about these opportunities so that arrangements can be made in advance.

For resource-poor countries, consider asking for donations from colleagues before leaving, including books, CDs, and medical supplies. Check local regulations first, particularly if bringing in medications or equipment. If sending books, some countries require high tariff fees to be paid by the receiver when accepting these packages, creating a financial burden for your hosts. Inquire ahead of time if they have to pay to accept these packages. Additionally, in some resource-poor countries, professionals do not have access to personal or work computers and internet café computers often do not have CD drives. Information on jump drives may be more easily accessible.

Finally, visiting educators may want to pack small gifts to give to hosts and others. These should be easily transported and may include items that represent your city or institution. We have also found bringing candy and small toys to be universally appreciated when visiting pediatric settings. A small portable color printer can be used to print photographs of pediatric patients as some of these children have never seen pictures of themselves. You can also print photographs of participants in the training courses.

Personal Considerations

Several months prior to departure, you should contact your traveler's health information resource to identify which vaccinations and what documents are needed to enter the country. To avoid lost time due to illness, ciprofloxacin and antidiarrheal medicines should be obtained before traveling. It is advisable to update your passport. Some countries require you to have sufficient blank pages in your passport to allow entry into their country. An entry fee paid in cash may be required upon arrival. Travelers should consider the political climate of the country and check the U.S. Department of State website (included in Table 1) for alerts or precautions.

Consider appropriate attire when packing. Clothing should reflect respect for the cultural and religious beliefs of the attendees.

During the Experience

It is very useful to meet with interpreters prior to the presentations to clarify any questions. Translation can be quite complicated. For example, a slide that used the term “caring” was interpreted as “romantic love,” and concepts about suffering and death can take on a cultural meaning. Check with interpreters regularly to determine if the speed of delivery is acceptable. Also, translators may have difficulty with this emotional content. In some instances, interpreters have become tearful and required debriefing after palliative care education events. Consider nonverbal communication and personal space. In some cultures, it may not be appropriate to shake hands or to use two hands. Gestures may have very different meanings in other cultures, so avoid these forms of communication. For example, the “OK” sign commonly used in North America, with the tip of the finger touching the tip of the thumb and the other three fingers extended, is considered an obscene gesture in Brazil.

When using teaching strategies other than lecture, respect that some students may not be comfortable at first with nontraditional approaches. Informal teaching strategies that are valued in North America may be viewed as of poor academic quality in other cultures. Debate and discussion, which may make it seem that the student is questioning a teacher's view, may be seen as disrespectful. At times, eliciting personal reflection and experience can engage the audience. For example, when introducing the topic of communication, health-care professionals in the audience can be asked the following questions:

• If you had cancer, would you want to know?

• How about your prognosis?

• Would you want to know that you had a disease that you could die from?

Following these with “What do you tell your patients?” usually engenders excellent discussion.

We have also found that asking participants to do “homework” can be useful, particularly if the students have been quiet or reluctant to communicate during class. Suggested assignments might include listing the five top barriers to cancer pain management in your setting, describing a difficult death or a death that you made better, or related issues. Reticence to speak during class may be due to discomfort with language skills. Some students feel more comfortable sharing ideas in writing, and these assignments have yielded valuable stories that have helped us to understand their experiences and perspectives.

Since the goal of these educational efforts should be sustained, it is helpful to develop a plan for the future with students. Assist them in identifying goals, as well as action items to meet these goals. Allow time for individual meetings between faculty and students to fine-tune these efforts. This ensures that the educational experience will have a greater likelihood of translation into action. To provide practical assistance, if Internet access is available, spend time with small groups to demonstrate literature searches, useful websites, and other information that will foster continuity.

Faculty should meet after each day of training to modify the planned agenda as needed, to optimally meet the needs of the participants. This also provides needed time to debrief about the day's activities and provide support. Particularly when new to international education, the experience may be overwhelming as the status of health-care in developing countries can cause deep personal reflection.

Finally, celebrate. We have found that many students appreciate the opportunity to have some type of closing ceremony to receive certificates and pins, acknowledge their accomplishments, and encourage their future efforts.

Afterward

E-mail, voiceover Internet services, and videoconferencing software have significantly enhanced global communication. Faculty can make themselves available to the trainees after leaving the country using these technologies. Group conversations via e-mail can help solve problems, provide encouragement, and celebrate successes. Connect attendees with international professional organizations to support ongoing educational efforts. It is very useful to identify the leaders or champions and to plan ongoing support to help sustain their commitment. Many countries do not have professional organizations or support networks. These leaders can exist in isolation and suffer great personal sacrifice to lead palliative care efforts in their country.

Conclusion

When educating about pain and palliative care to a worldwide audience, never make assumptions, expect the unexpected, and be flexible. We have found many of these international teaching experiences to be some of the most exhilarating of our professional lives, providing insight to our own practices and creating lasting relationships with colleagues from around the globe. Ultimately, these efforts will improve care for people with cancer.

 

 

Acknowledgments

The authors acknowledge the American Association of Colleges of Nursing and the City of Hope for their ongoing support of the End-of-Life Nursing Education Consortium training activities, as well as the Oncology Nursing Society Foundation and the Open Society Institute for their support of international educational efforts. They also thank Marian Grant for her input.

References [Pub Med ID in Brackets]

1 A.L. Taylor, L.O. Gostin and K.A. Pagonis, Ensuring effective pain treatment: a national and global perspective, JAMA 299 (2008), pp. 89–91 [18167410]. 

2 K. Crane, Palliative care gains ground in developing countries, J Natl Cancer Inst 102 (21) (2010), pp. 1613–1615 [20966432]. 

3 J.A. Paice, B.R. Ferrell, N. Coyle, P. Coyne and M. Callaway, Global efforts to improve palliative care: the International End-of-Life Nursing Education Consortium training programme, J Adv Nurs 61 (2007), pp. 173–180 [18186909].

4 J.A. Paice, B. Ferrell, N. Coyle, P. Coyne and T. Smith, Living and dying in East Africa: implementing the End-of-Life Nursing Education Consortium curriculum in Tanzania, Clin J Oncol Nurs 14 (2010), pp. 161–166 [20350889]. 

5 C. Olweny, C. Sepulveda, A. Merriman, S. Fonn, M. Borok, T. Ngoma, A. Doh and J. Stjernsward, Desirable services and guidelines for the treatment and palliative care of HIV disease patients with cancer in Africa: a World Health Organization consultation, J Palliat Care 19 (2003), pp. 198–205 [14606333]. 

6 C. Sepulveda, V. Habiyatmbete, J. Amandua, M. Borok, E. Kikule, B. Mudanga and B. Solomon, Quality care at the end of life in Africa, BMJ 327 (2003), pp. 209–213 [12881267]. 

7 E.L. Krakauer, R. Wenk, R. Buitrago, P. Jenkins and W. Scholten, Opioid inaccessibility and its human consequences: reports from the field, J Pain Palliat Care Pharmacother 24 (2010), pp. 239–243 [20718644].

8 C.M. Bolin, Developing a postbasic gerontology program for international learners: considerations for the process, J Contin Educ Nurs 34 (2003), pp. 177–183 [12887229].

9 K.D. Meneses and C.H. Yarbro, Cultural perspectives of international breast health and breast cancer education, J Nurs Scholarsh 39 (2) (2007), pp. 105–112 [19058079]. 

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and none were reported.


Correspondence to: Judith A. Paice, PhD, RN, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair Street, Suite 850, Chicago, IL 60611; telephone: (312) 695-4157; fax: (312) 695-6189.

 


Vitae

Dr. Paice is Director of the Cancer Pain Program, Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.

Carma Erickson-Hurt is a faculty member at Grand Canyon University, Phoenix, Arizona.

Dr. Ferrell is a Professor and Research Scientist at the City of Hope National Medical Center, Duarte, California.

Nessa Coyle is on the Pain and Palliative Care Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York.

Dr. Coyne is Clinical Director of the Thomas Palliative Care Program, Virginia Commonwealth University/Massey Cancer Center, Richmond, Virginia.

Dr. Long is a geriatric nursing consultant and codirector of the Palliative Care for Advanced Dementia, Beatitudes Campus, Phoenix, Arizona.

Dr. Mazanec is a clinical nurse specialist at the University Hospitals Seidman Cancer Center, Cleveland, Ohio.

Pam Malloy is ELNEC Project Director, American Association of Colleges of Nursing, Washington, DC.

Dr. Smith is Professor of Medicine and Palliative Care Research, Virginia Commonwealth University/Massey Cancer Center, Richmond.

Publications
Publications
Article Type
Display Headline
Providing Pain and Palliative Care Education Internationally
Display Headline
Providing Pain and Palliative Care Education Internationally
Legacy Keywords
Pain, Palliative Care, Cancer, International, Education
Legacy Keywords
Pain, Palliative Care, Cancer, International, Education
Sections
Disallow All Ads
Vitals

For many clinicians in oncology, educating other health-care professionals about cancer pain and palliative care is part of their professional life. The need for education exists across clinical settings around the world. Improved education is an urgent need as the prevalence of cancer is increasing.

 

 

 

Facilitating hospice discussions: a six-step roadmap

Article Type
Changed
Fri, 01/04/2019 - 11:03
Display Headline
Facilitating hospice discussions: a six-step roadmap

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

 

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

 

Table 1. Hospice Services and Team Members

 

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

 

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

 

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

 

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

 

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

 

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

 

 

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

 

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

 

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

 

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

 

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

 

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

 

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

 

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

 

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104

 


1 PubMed ID in brackets

 

 

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia

 

 


 

 

Author and Disclosure Information

 

 

Publications
Topics
Sections
Author and Disclosure Information

 

 

Author and Disclosure Information

 

 

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

 

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

 

Table 1. Hospice Services and Team Members

 

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

 

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

 

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

 

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

 

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

 

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

 

 

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

 

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

 

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

 

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

 

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

 

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

 

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

 

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

 

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104

 


1 PubMed ID in brackets

 

 

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia

 

 


 

 

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

 

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

 

Table 1. Hospice Services and Team Members

 

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

 

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

 

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

 

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

 

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

 

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

 

 

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

 

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

 

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

 

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

 

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

 

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

 

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

 

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

 

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104

 


1 PubMed ID in brackets

 

 

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia

 

 


 

 

Publications
Publications
Topics
Article Type
Display Headline
Facilitating hospice discussions: a six-step roadmap
Display Headline
Facilitating hospice discussions: a six-step roadmap
Sections
Disallow All Ads
Alternative CME

Facilitating Hospice Discussions: A Six-Step Roadmap

Article Type
Changed
Fri, 01/04/2019 - 11:02
Display Headline
Facilitating Hospice Discussions: A Six-Step Roadmap

How we do it

Facilitating Hospice Discussions: A Six-Step Roadmap

Jennifer Shin MD

,
and David Casarett MD, MA   [Author vitae]

Received 28 December 2010; 

accepted 18 March 2011. 

Available online 10 May 2011.

Abstract

Hospice programs provide comprehensive, compassionate care to dying patients and their families. However, many patients do not enroll in hospice, and those who do generally receive hospice care only in the last weeks of life. Although patients and families rely on their physicians to discuss hospice, there is often inadequate communication between patients and physicians about end-of-life issues. We describe a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news: setting up the discussion, assessing the patient’s perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient’s emotions, and summarizing and strategizing the next steps.

Article Outline

Case

Introduction

When Is a Hospice Discussion Appropriate?

A Six-Step Roadmap
Step 1: Set Up the Discussion About Hospice
Step 2: Assess the Patient's Perception
Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care
Step 4: Share Knowledge
Step 5: Empathize With the Patient's Emotion
Step 6: Summarize the Discussion and Strategize Next Steps

Conclusion

References

Vitae

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

Table 1. Hospice Services and Team Members

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104


1 PubMed ID in brackets

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia


Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

How we do it

Facilitating Hospice Discussions: A Six-Step Roadmap

Jennifer Shin MD

,
and David Casarett MD, MA   [Author vitae]

Received 28 December 2010; 

accepted 18 March 2011. 

Available online 10 May 2011.

Abstract

Hospice programs provide comprehensive, compassionate care to dying patients and their families. However, many patients do not enroll in hospice, and those who do generally receive hospice care only in the last weeks of life. Although patients and families rely on their physicians to discuss hospice, there is often inadequate communication between patients and physicians about end-of-life issues. We describe a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news: setting up the discussion, assessing the patient’s perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient’s emotions, and summarizing and strategizing the next steps.

Article Outline

Case

Introduction

When Is a Hospice Discussion Appropriate?

A Six-Step Roadmap
Step 1: Set Up the Discussion About Hospice
Step 2: Assess the Patient's Perception
Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care
Step 4: Share Knowledge
Step 5: Empathize With the Patient's Emotion
Step 6: Summarize the Discussion and Strategize Next Steps

Conclusion

References

Vitae

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

Table 1. Hospice Services and Team Members

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104


1 PubMed ID in brackets

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia


How we do it

Facilitating Hospice Discussions: A Six-Step Roadmap

Jennifer Shin MD

,
and David Casarett MD, MA   [Author vitae]

Received 28 December 2010; 

accepted 18 March 2011. 

Available online 10 May 2011.

Abstract

Hospice programs provide comprehensive, compassionate care to dying patients and their families. However, many patients do not enroll in hospice, and those who do generally receive hospice care only in the last weeks of life. Although patients and families rely on their physicians to discuss hospice, there is often inadequate communication between patients and physicians about end-of-life issues. We describe a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news: setting up the discussion, assessing the patient’s perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient’s emotions, and summarizing and strategizing the next steps.

Article Outline

Case

Introduction

When Is a Hospice Discussion Appropriate?

A Six-Step Roadmap
Step 1: Set Up the Discussion About Hospice
Step 2: Assess the Patient's Perception
Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care
Step 4: Share Knowledge
Step 5: Empathize With the Patient's Emotion
Step 6: Summarize the Discussion and Strategize Next Steps

Conclusion

References

Vitae

Case

Mr. C is a 54-year-old man with metastatic lung adenocarcinoma who presented 1 year ago with multiple lung nodules and hepatic metastases. His tumor responded to initial treatment with four cycles of carboplatin and pemetrexed, and he was treated with maintenance pemetrexed for 5 months before his liver lesions progressed. Despite treatment with docetaxel and then erlotinib, his liver and lung tumors progressed rapidly. He has lost 10 pounds in the last few weeks and is now so fatigued that he spends most of his day resting in his recliner. He has been admitted to the hospital twice in the last month for worsening dyspnea in the setting of progressive pulmonary metastases.

Introduction

Hospice programs provide high-quality, compassionate care to dying patients and their families through an interdisciplinary team specializing in pain and symptom management.[1] and [2] These patients also receive medications and supplies, durable medical equipment, and home health aide services. Patients and their families receive support from a chaplain, social worker, physician, nurse, and volunteer. Families are offered bereavement services for at least 13 months following the patient's death (Table 1).3

Table 1. Hospice Services and Team Members

Services
• Pain and symptom management
• 24-hour telephone access to a clinician
• Assistance with personal care needs
• Help with errands and light housework
• Spiritual support
• Companionship for the patient and family
• Bereavement counseling before and after the patient's death
• Patient and family education and counseling
• Case management and coordination
• Advance care planning
• Medications and supplies related to the hospice diagnosis
• Durable medical equipment
• Child bereavement services
• Respite services (up to 5 consecutive days of inpatient care to allow families a needed break)
• Inpatient hospice (for treatment of severe symptoms that cannot be managed at home)
Team members
• Physician
• Nurse
• Social worker
• Home health aide
• Chaplain
• Bereavement counselor
• Physical therapist
• Occupational therapist
• Volunteer

National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America.3

Families report high levels of satisfaction with hospice and are more likely to describe high-quality care.1 Despite this, only 40% of people in the United States die while receiving hospice care.3 Although longer hospice stays are associated with better quality of life in patients and less depression in bereaved family members,[4] and [5] many patients enroll very late in the course of illness. In fact, the median length of stay is just over 3 weeks, and a third of patients die or are discharged within 1 week of hospice enrollment.3 The result is that brief exposures to hospice at the end of life do not allow patients and families to take full advantage of the benefits.6

There are several explanations for why people enroll in hospice so late. The Medicare Hospice Benefit requires hospice patients to choose a plan of comfort care, which means that they must usually forgo disease-directed therapies unless they provide a specific palliative benefit. Furthermore, the financial per diem payment structure of hospice means that patients may need to forgo palliative treatments that cannot be covered under the typical hospice reimbursement (approximately $150/day for routine home care). Patients may not be willing to give up these therapies or may be reluctant to transition from a model of care focusing on disease-directed therapies to one with palliation as the goal.

Patients may also enroll in hospice later if their physicians do not discuss hospice or if they have these discussions in the last few weeks of the patient's life.[7] and [8] Physicians may delay hospice discussions because they are unsure of the patient's prognosis,9 although the disease trajectory in patients with cancer is often more straightforward than in patients with non-oncologic diagnoses. Studies have documented deficiencies in doctor–patient communication regarding prognosis and end-of-life issues,10 and patients report inadequate communication with physicians about shared decision making at the end of life.1 Another study found that about half of patients diagnosed with metastatic lung cancer reported not having discussed hospice with a provider within 4–7 months after diagnosis.11

Although these discussions may be delayed or avoided altogether, seriously ill patients value the ability to prepare for the end of life.[12] and [13] Patients rely on their physicians to discuss hospice and other end-of-life care options. Furthermore, most family caregivers report that communication with their oncologists was important in helping them to understand the patient's prognosis and to see the role that hospice could play as a treatment alternative.7

These conversations are often difficult for patients and families and can also be challenging for physicians. Nevertheless, communication skills in discussing transitions to palliative care can be learned.[14] and [15] Although these discussions are not nearly as straightforward as a medical or surgical procedure, one can approach them with the same methodical preparation and careful consideration of the steps involved.

When Is a Hospice Discussion Appropriate?

To be eligible for the Medicare Hospice Benefit, a patient must have a prognosis of 6 months or less if his or her illness runs its usual course; also, the patient needs to be willing to accept the hospice philosophy of comfort care. This second criterion is not formally defined but is generally accepted to mean that the patient must be willing to forgo disease-directed therapies related to the hospice admitting diagnosis.

These eligibility criteria should not be used to define the patients for whom a hospice discussion is appropriate however. When a patient's goals and values reflect a desire to focus on palliation, it is time for the physician to initiate a hospice discussion. Other triggers for early hospice discussions can include a change in clinical status, recent hospitalization, decline in performance status, new weight loss, or complication of treatment. Although these factors may prompt a discussion of options for care, including hospice,[16] and [17] not all discussions will lead to a hospice enrollment decision. Nevertheless, earlier discussions that prompt conversations about a patient's needs, goals, and preferences can facilitate later decisions about hospice and other treatment options.

A Six-Step Roadmap

We provide a Six-Step Roadmap for navigating discussions about hospice adapted from the SPIKES protocol for delivering bad news.18 This strategy is comprised of six communication steps that can be remembered by using the mnemonic SPIKES: setting up the discussion, assessing the patient's perception, inviting a patient to discuss individual goals and needs, sharing knowledge, empathizing with the patient's emotions, and summarizing and strategizing the next steps.

Step 1: Set Up the Discussion About Hospice

Before discussing hospice with a patient and family, it is important to communicate with other members of the medical team to ensure an understanding of the patient's prognosis and treatment options. It is also helpful to find out what the patient and family may have expressed to other providers regarding these issues and how they have been coping. Any provider who has been in contact with the patient may be able to contribute to this consensus, including the medical oncologist, radiation oncologist, palliative care physician, primary care physician, home nurse, and social worker. A clear, unified message from the team decreases confusion for the patient and family.

Once a common agreement has been established regarding the patient's prognosis and treatment options, physicians can schedule a time and arrange for a place to allow for an uninterrupted conversation. Scheduling a patient at the end of a clinic day or visiting a patient in the hospital during an admission are potential ways to do this. Before scheduling a meeting, however, it is essential to know who the patient would like to be present at the meeting. One approach may be to tell the patient that there are important options to discuss regarding the next steps in his or her care and find out who may be able to help the patient with such decisions (Table 2). Additionally, a palliative care physician may cofacilitate these discussions. If the patient already has a palliative care physician, it may be helpful to have him or her involved in the meeting. If the patient has not yet been evaluated by a palliative care team, it may be possible to consult a palliative care specialist who can attend the meeting or follow up with the patient afterward.

 

 

Table 2. Useful Language for Hospice Discussions

Adapted from Cassett et al8 and Baille et al15

Invite other decision makers“Who do you usually rely on to help you make important decisions?”
“When we discuss your results, who would you like to be present?”
Assess understanding of prognosis“Tell me about your understanding of the most recent tests/studies.”
“Can you share with me what you think is happening with your cancer and the treatments?”
Identify goals of care“What is most important to you right now?”
“What are your biggest concerns right now?”
“What are your hopes for the coming weeks/months?”
“What do you enjoy doing now?”
“What is most important to you now?”
“What are you worried about now? In the future?”
Reframe goals (“wish” statements)“I wish I could promise you that you will be able to make it to your daughter's wedding, but unfortunately I can't. What do you think about writing a letter for her to read on her wedding day? We can also think about other ways to let her know that you will always be with her, even if you cannot physically be there.”
“I wish that we could find a new chemotherapy that could cure your cancer. Even though cure is not possible, I think that we can meet some of your other goals, like staying at home to spend time with your children.”
Identify needs for care“What has been hard for you and your family?”
“What is your life like when you are at home? How are you and your family managing?”
“Are you experiencing pain or other symptoms that are bothering you?”
“Have you been feeling sad or anxious lately?”
“Would it be helpful to have a visiting nurse come to your home to assist you with your medications?”
Introduce hospice“One of the best ways to give you the help that you need to stay at home is through hospice.”
“The hospice team specializes in caring for seriously ill patients at home.”
“Hospice can provide you and your family with more services and support.”
Recommend hospice“From what you have shared with me today, I recommend hospice as a way of helping you meet the goals that are important to you.”
“I feel that hospice is the best option for you and your family. I know this is a big decision, and I want you to know that the decision is yours.”

Mr. C's medical oncologist, Dr. A, contacted Mr. C's radiation oncologist as well as his primary care physician. They all agreed that his prognosis could be measured in weeks to months and that his performance status precluded any further chemotherapy. Dr. A also spoke with the hospital social worker who met Mr. C and his wife during his most recent hospitalization. The social worker said that Mr. C's wife has been very distressed, particularly about his increasing debility and her difficulty in caring for him at home. When Dr. A visited Mr. C during his hospitalization, Dr. A explained to Mr. C that they would be making some decisions about the next steps in his care and asked who might be able to help with these decisions. When Mr. C said his wife would be this person, Dr. A asked that she come to his next visit. Dr. A decided to schedule Mr. C for an appointment at the end of his clinic session the following week.

Step 2: Assess the Patient's Perception

The physician can begin this discussion by asking the patient to describe his or her current medical situation (Table 2). Although the physician may have provided this information on prior occasions, it is important to hear the patient's perception of the diagnosis and prognosis. Patients with advanced cancer often overestimate their prognosis and are more likely to favor life-extending therapies over hospice.19 These questions provide an opportunity to address any misconceptions or gaps in understanding that the patient may have. When the physician, patient, and family are in agreement with the patient's current medical situation it allows for further exploration of the patient's hopes and concerns.

This part of the discussion should rely on open-ended questions designed to elicit the patient's perspective. In particular, an invitation to “tell me more” encourages patients to explore how they are thinking or feeling and can yield more information than closed-ended or leading questions. This phrase can also help redirect the conversation when necessary (“You mentioned before that you are worried that the chemotherapy is not working anymore. Tell me more about your concerns.”).

Dr. A asked Mr. C how he was doing overall and to describe his understanding of whether the erlotinib had been working. In response, Mr. C expressed his concern about his recent weight loss and lethargy. Dr. A asked Mr. C to tell him more about these concerns, and Mr. C said that he thought his symptoms were a sign that the erlotinib was not helping him. He said he knew that the CT scans showed progression of disease, and he wondered whether chemotherapy could help. Dr. A confirmed there was progression of cancer in his lungs and liver. Dr. A also expressed his concern that more chemotherapy would not provide additional benefit for him and may harm him. Mr. C and his wife were tearful and agreed that he was too weak for more chemotherapy. Dr. A acknowledged that the disease had progressed quickly and must be very upsetting to them.

Step 3: Invite the Patient to Discuss Goals of Care and Needs for Care

Before sharing information about hospice with a patient, it is important to understand the patient's hopes and fears about the future, goals of care, and needs for care. It is helpful to start with learning about the patient's perspectives on the future and linking that to the patient's goals of care. Once the goals are clear, it is easier to match the patient's needs with his or her goals.

One way to elicit patients' goals of care is by asking them to describe their hopes and fears about their cancer in the context of their life (Table 2). Patients may volunteer information about their hopes (eg, attending their daughter's wedding) or fears (eg, worrying about pain) that provide insight into their more global goals of care. Again, the “tell me more” phrase can be helpful (“Tell me more about what you mean when you say you are a burden on your family.”).

Once the patient and family express their thoughts, it is useful to restate the patient's goals by asking a question that summarizes the patient's statements (“From what you and your family have just shared with me, I hear that the most important thing to you is … . Did I understand you correctly?”). It is often challenging for patients to specifically articulate their goals of care. Asking a question allows the patient and family the chance to elaborate or offer corrections.

If a patient expresses unrealistic expectations (eg, a cure, years of life), “wish” statements can be helpful in providing gentle redirection. These statements express empathy while also communicating that the wished-for outcome is unlikely (“I wish that we could guarantee that … but unfortunately we can't.”). These statements can explain the reality of the situation in a compassionate manner (Table 2). Patients and families who have unrealistic goals of care may need time to readjust their expectations, and in these cases it may be prudent to revisit the discussion of hospice at a later date.

Once the goals of care have been established, it is important to further explore the needs for care. Although some of these needs may have been mentioned during the goals discussion, it is helpful to directly ask the patient and family about their needs. General questions about what has been hard for the patient and the family can be useful in eliciting needs, as are questions about what life has been like at home and how they are managing (Table 2). It is also important to ask more specific questions that pertain to the patient's symptoms such as pain or depression and those that address the family's needs for help around the house (Table 2). Once this information has been shared, it is often useful to repeat a summary back to the patient and family (“From what we have just discussed, it may be helpful to have a visiting nurse to assist with his medications and a home health aide to dress and bathe him … . Does it sound like this could be helpful to you?”). Often, these needs can be addressed by the multidisciplinary hospice team, and it is important to understand what needs exist in preparation for a discussion about how hospice might be helpful.

Mr. C shares with you that his two hospitalizations for dyspnea have been frustrating because he feels that they have prevented him from spending quality time with his daughters. Although he did not want to be admitted to the hospital, he tearfully expressed that he was worried about “suffocating to death” and did not want to die at home in front of his wife and children. Mr. C's wife also shares that it has been harder to bathe and dress him because he is becoming so weak. Mr. and Mrs. C agree that it would be helpful to have the support of a visiting nurse and a home health aide.

Step 4: Share Knowledge

Once the patient's goals and needs for care have been clarified, physicians can introduce hospice as a way of achieving their goals and meeting their needs. In presenting hospice in this transparent manner, patients and families can better understand how hospice is part of a plan of care that addresses their individualized goals and needs. Most family caregivers report that communication with their oncologists was critical in their understanding the patient's prognosis and hospice as a treatment alternative.7 In one survey, the majority of caregivers did not realize that their loved ones could benefit from hospice until their physicians first discussed it with them.7

A discussion of hospice should offer concrete information about the services provided to patients and their families (Table 1). Many patients and families do not understand the benefits, such as a visiting nurse for frequent symptom management or a home health aide to assist with daily patient care, until after enrollment. Many say they wish they had known sooner.20 By providing this information earlier, patients and families may make more informed decisions about hospice. This description also makes clear to the patient and family that hospice is not simply a generic recommendation but rather the physician's recommendation of a program that is the best fit for their specific goals and needs.

Given the emotional nature of these discussions and the large amount of information involved, it is important to ask the patient to explain in his or her own words how hospice could help (“To make sure I did a good job of explaining things, can you tell me what we just talked about in your own words?” and “How do you think hospice might help you?”). This provides the opportunity to assess understanding and clarify any confusion.

Since not all patients are best served by hospice, the discussion may also be expanded to include other options for palliative care. For example, hospice is not equipped to care for debilitated patients at home who do not have a caretaker, nor is it usually able to absorb the costs of expensive palliative treatments. Sometimes, larger hospices may be able to make exceptions on a case-by-case basis, but it is helpful to be aware of other options for palliative care like bridge-to-hospice home care and outpatient palliative care programs.

Dr. A explained that hospice could provide intensive management of his dyspnea with the assistance of a visiting nurse and a 24-hour phone line to call for assistance. With these measures in place, Dr. A said that he hoped hospitalizations could be avoided, allowing him more time at home with his family. Dr. A addressed Mr. C's concern about dying at home and shared with him that he could be transferred to an inpatient hospice if death seemed imminent. Dr. A shared with Mrs. C that hospice could provide the services of a home health aide, which seemed to reduce her concerns about being able to care for her husband as he became weaker.

Step 5: Empathize With the Patient's Emotion

In discussions regarding end-of-life care, patients and families value empathy, compassion, and honesty balanced with sensitivity and hope.21 Throughout the conversation, it is likely that the patient and family will express a range of emotions. Rather than providing immediate reassurance or trying to “fix” the emotion, it can be helpful to use an empathic statement to let the patient know that his or her emotions are recognized.15 Empathic responses address and validate a patient's emotions and encourage further disclosure.22 The NURSE mnemonic summarizes ways in which to respond to emotions: naming, understanding, respecting, supporting, and exploring the feelings the patient has shared (Table 3).[15] and [23]

 

 

Table 3. NURSE Statements for Expressing Empathy

Adapted from Back et al[15] and [23]

N = Naming“It sounds like you are worried about how fast the cancer has been progressing.”
“Some people in this situation would feel frustrated.”
U = Understanding“My understanding of what you have told me is that you are worried about being able to live independently at home.”
“I can see how difficult this has been for you and your family.”
R = Respecting“It is very clear to me how supportive your family has been.”
“I can see how hard you have worked to understand the treatment options for your cancer.”
S = Supporting“I will support the decisions that you make, no matter what you decide.”
“I will always be your doctor.”
E = Exploring“Could you tell me more about what you mean when you say that you don't want to give up?”
“I sense that you may be feeling anxious about stopping chemotherapy. Can you share with me what you are feeling?”

Some of the emotions arising during a hospice discussion may stem from preconceived notions or a prior experience. Therefore, it may be helpful to specifically ask patients and families about these perceptions and experiences. Common misperceptions may include the concern that hospice hastens death. Other patients view hospice as “giving up” and worry about being abandoned by their physicians. A hospice discussion provides the opportunity to directly address these concerns and provide clarification (“No, hospice does not hasten death. Hospice helps you have the best quality of life for whatever time you have.”).

Ultimately, some patients and families may decide that hospice is not the right choice for them. It is important to recognize that the time invested has not been wasted. Instead, if done well, these discussions offer an opportunity for the physician to align his or her goals and understanding with those of the patient and family. Specifically, these discussions are a chance to demonstrate a desire to understand the patient's individualized goals and to share concerns about disease progression. In essence, it is a valuable opportunity to establish a collective understanding about the patient's current situation while also laying important groundwork for future discussions.

Mr. C tearfully shared his concern about being a burden to his wife and about how his daughters would handle his progressive decline. Dr. A sat quietly, allowed Mr. C to fully detail his worries, and then said, “I can see how worried you are about your family and understand that you want to make sure that their needs are also addressed.”

Step 6: Summarize the Discussion and Strategize Next Steps

In all stages of cancer, patients and families rely on their oncologist for information about treatment options. This is particularly important when a patient's cancer has progressed despite therapy and when the focus of care may be shifting from disease-directed therapies to palliation. Just as a physician may have previously recommended a chemotherapeutic option for a patient, so should he or she recommend the therapeutic option of focusing on quality of life. If hospice appears consistent with the patient's and family's goals and needs, the physician should make this recommendation. It may be helpful for the patient and family to hear a summary of how hospice will meet their needs. If the patient is amenable to learning more about hospice but is not yet ready to enroll, the physician can arrange for an informational visit with the hospice team.

Dr. A recommends hospice and emphasizes that hospice would provide services that meet Mr. C's goals of symptom management, avoiding hospitalizations, and providing support for his family. Mr. C and his wife agree that hospice is the best option for them. They would like to enroll after they have spoken with their children about their decision.

Conclusion

Discussions about goals of care and hospice are not easy. They are rarely as straightforward as presented in this case, and an oncologist may face numerous barriers when attempting to have these discussions. For instance, treating physicians may have differing opinions on therapeutic options. Patients and family members may have different goals and may be in different stages of accepting a life-limiting cancer diagnosis. Additionally, these discussions take preparation, time, and skill. Although there are no easy solutions to these issues, the general guidance provided in this article focuses on suggesting tools and techniques that can make these discussions easier for oncologists, patients, and families. By increasing our competence and comfort with these conversations, we can reduce delays in offering patients the benefits of hospice as they near the end of life.

Although no algorithm will fully address the complexities and nuances of these conversations, this approach provides a general framework and offers tools to use while speaking with patients and families. Conversations about hospice do not begin with the recommendation of hospice but rather with an honest discussion of the goals and needs of a patient and family. If these goals and needs can be met with the services that hospice can provide, the physician has the opportunity to educate the patient and to make the recommendation as they would for any other therapeutic option.

Patients and families consider communication to be one of the most important facets of end-of-life care.24 Seriously ill patients value being able to prepare for death,13 and physicians have the duty to help patients and families prepare for the end of life.25 Physicians can help increase the time patients have to plan for the last phase of their lives by having honest and open discussions about hospice and other alternatives. Oncologists have the responsibility to present patients with the benefits and burdens of therapies throughout the trajectory of their illness, and it is critically important during the transition from disease-directed to palliative care. By exploring the option of hospice, patients and families can make informed decisions about whether hospice may meet their needs. Equally important, patients are given the control to choose how they would like to live the final phase of their lives.

 

 

References1

1 J.M. Teno, B.R. Clarridge, V. Casey, L.C. Welch, T. Wetle, R. Shield and V. Mor, Family perspectives on end-of-life care at the last place of care, JAMA 291 (1) (2004), pp. 88–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (411)

2 S.C. Miller, V. Mor and J. Teno, Hospice enrollment and pain assessment and management in nursing homes, J Pain Symptom Manage 26 (3) (2003), pp. 791–799. Article |

PDF (106 K)
| View Record in Scopus | Cited By in Scopus (67)

3 National Hospice and Palliative Care Organization, NHPCO Facts and Figures: Hospice Care in America www.nhpco.org/files/public/Statistics_Research/Hospice_Facts_Figures_Oct-2010.pdf+NHPCO+Facts+and+Figures:+Hospice+Care+in+America.+National+Hospice+and+Palliative+Care+Organization&hl=en&gl=us&pid=bl&srcid=ADGEESgPEeCPa2C7KZ3zQT0yldBWHrCAHY4UemVw6R1Odl2VxMKsjrMnNridOO3qiHrvN9cAfwCzkIY3Mc28JRgBLdvCaN3nQXt7EUNL6H-PTkYYaVAl4-VECDAsh16DvmofChWiLRfa&sig=AHIEtbTMY3LallQ5V2Ceyd_ACIQfUh-ggA (2010).

4 A.A. Wright, B. Zhang, A. Ray, J.W. Mack, E. Trice, T. Balboni, S.L. Mitchell, V.A. Jackson, S.D. Block, P.K. Maciejewski and H.G. Prigerson, Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment, JAMA 300 (14) (2008), pp. 1665–1673. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (131)

5 E.H. Bradley, H. Prigerson, M.D.A. Carlson, E. Cherlin, R. Johnson-Hurzeler and S.V. Kasl, Depression among surviving caregivers: does length of hospice enrollment matter?, Am J Psychiatry 161 (12) (2004), pp. 2257–2262. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

6 I.R. Byock, W.B. Forman and M. Appleton, Academy of hospice physicians' position statement on access to hospice and palliative care, J Pain Symptom Manage 11 (2) (1996), pp. 69–70. Article |

PDF (131 K)
| View Record in Scopus | Cited By in Scopus (17)

7 E. Cherlin, T. Fried, H.G. Prigerson, D. Schulman-Green, R. Johnson-Hurzeler and E.H. Bradley, Communication between physicians and family caregivers about care at the end of life: when do discussions occur and what is said?, J Palliat Med 8 (6) (2005), pp. 1176–1185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)

8 D.J. Casarett and T.E. Quill, “I'm not ready for hospice”: strategies for timely and effective hospice discussions, Ann Intern Med 146 (6) (2007), pp. 443–449. View Record in Scopus | Cited By in Scopus (36)

9 E.B. Lamont and N.A. Christakis, Prognostic disclosure to patients with cancer near the end of life, Ann Intern Med 134 (12) (2001), pp. 1096–1105. View Record in Scopus | Cited By in Scopus (166)

10 M. Gysels, A. Richardson and I.J. Higginson, Communication training for health professionals who care for patients with cancer: a systematic review of effectiveness, Support Care Cancer 12 (10) (2004), pp. 692–700. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)

11 H.A. Huskamp, N.L. Keating, J.L. Malin, A.M. Zaslavsky, J.C. Weeks, C.C. Earle, J.M. Teno, B.A. Virnig, K.L. Kahn, Y. He and J.Z. Ayanian, Discussions with physicians about hospice among patients with metastatic lung cancer, Arch Intern Med 169 (10) (2009), pp. 954–962. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

12 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, S. Grambow, J. Parker and J.A. Tulsky, Preparing for the end of life: preferences of patients, families, physicians, and other care providers, J Pain Symptom Manage 22 (3) (2001), pp. 727–737. Article |

PDF (78 K)
| View Record in Scopus | Cited By in Scopus (120)

13 K.E. Steinhauser, N.A. Christakis, E.C. Clipp, M. McNeilly, L. McIntyre and J.A. Tulsky, Factors considered important at the end of life by patients, family, physicians, and other care providers, JAMA 284 (19) (2000), pp. 2476–2482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (615)

14 A.L. Back, R.M. Arnold, W.F. Baile, K.A. Fryer-Edwards, S.C. Alexander, G.E. Barley, T.A. Gooley and J.A. Tulsky, Efficacy of communication skills training for giving bad news and discussing transitions to palliative care, Arch Intern Med 167 (5) (2007), pp. 453–460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (94)

15 A.L. Back, R.M. Arnold, W.F. Baile, J.A. Tulsky and K. Fryer-Edwards, Approaching difficult communication tasks in oncology, CA Cancer J Clin 55 (3) (2005), pp. 164–177. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (103)

16 C. Conill, E. Verger and M. Salamero, Performance status assessment in cancer patients, Cancer 65 (8) (1990), pp. 1864–1866. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)

17 V. Mor, L. Laliberte, J.N. Morris and M. Wiemann, The Karnofsky Performance Status Scale: An examination of its reliability and validity in a research setting, Cancer 53 (9) (1984), pp. 2002–2007. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (314)

18 W.F. Baile, R. Buckman, R. Lenzi, G. Glober, E.A. Beale and A.P. Kudelka, SPIKES—a six-step protocol for delivering bad news: application to the patient with cancer, Oncologist 5 (4) (2000), pp. 302–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (293)

19 J.C. Weeks, E.F. Cook, S.J. O'Day, L.M. Peterson, N. Wenger, D. Reding, F.E. Harrell, P. Kussin, N.V. Dawson, A.F. Connors, J. Lynn and R.S. Phillips, Relationship between cancer patients' predictions of prognosis and their treatment preferences, JAMA 279 (21) (1998), pp. 1709–1714. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (350)

20 D.J. Casarett, R.L. Crowley and K.B. Hirschman, How should clinicians describe hospice to patients and families?, J Am Geriatr Soc 52 (11) (2004), pp. 1923–1928. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)

21 S.M. Parker, J.M. Clayton, K. Hancock, S. Walder, P.N. Butow, S. Carrick, D. Currow, D. Ghersi, P. Glare, R. Hagerty and M.H. Tattersall, A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information, J Pain Symptom Manage 34 (1) (2007), pp. 81–93. Article |

PDF (166 K)
| View Record in Scopus | Cited By in Scopus (44)

22 K.I. Pollak, R.M. Arnold, A.S. Jeffreys, S.C. Alexander, M.K. Olsen, A.P. Abernethy, C. Sugg Skinner, K.L. Rodriguez and J.A. Tulsky, Oncologist communication about emotion during visits with patients with advanced cancer, J Clin Oncol 25 (36) (2007), pp. 5748–5752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)

23 A.L. Back, W.G. Anderson, L. Bunch, L.A. Marr, J.A. Wallace, H.B. Yang and R.M. Arnold, Communication about cancer near the end of life, Cancer 113 (suppl 7) (2008), pp. 1897–1910. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)

24 M.D. Wenrich, J.R. Curtis, S.E. Shannon, J.D. Carline, D.M. Ambrozy and P.G. Ramsey, Communicating with dying patients within the spectrum of medical care from terminal diagnosis to death, Arch Intern Med 161 (6) (2001), pp. 868–874. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (114)

25 J. Lynn, Perspectives on care at the close of life: Serving patients who may die soon and their families: the role of hospice and other services, JAMA 285 (7) (2001), pp. 925–932. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (124)

 

 

Dr. Casarett's research is funded by grants from the Commonwealth Fund, the Greenwall Foundation, the VistaCare Foundation, and the Department of Veterans' Affairs. Dr. Shin has nothing to disclose.

Correspondence to: Jennifer Shin, MD, Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104


1 PubMed ID in brackets

Vitae

Dr. Shin is from the Division of Hematology–Oncology, University of Pennsylvania School of Medicine, Philadelphia

Dr. Casarett is from the Division of Geriatric Medicine and the Penn-Wissahickon Hospice, University of Pennsylvania School of Medicine, Philadelphia


Publications
Publications
Topics
Article Type
Display Headline
Facilitating Hospice Discussions: A Six-Step Roadmap
Display Headline
Facilitating Hospice Discussions: A Six-Step Roadmap
Sections
Article Source

PURLs Copyright

Inside the Article

Recognizing Depression in Patients with Cancer

Article Type
Changed
Fri, 01/04/2019 - 11:02
Display Headline
Recognizing Depression in Patients with Cancer

How we do it

Recognizing Depression in Patients with Cancer

Alicia Morgans MD
,
and Lidia Schapira MD

Received 2 August 2010; 

accepted 20 December 2010. 

Available online 2 April 2011.

Article Outline

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

Should the Oncologist Offer Treatment for Depression?

Conclusion

Acknowledgements

References

Caring for the “whole person” across the continuum of disease remains a top priority for clinicians. Strides in basic and translational research have opened new therapeutic pathways that provide better targeted and effective treatments with fewer side effects. New ancillary and supportive therapies have transformed and improved the experiences of patients undergoing anticancer treatments. Also, with increasing acceptance of multidisciplinary teams, we have the opportunity to make timely referrals to colleagues who provide supportive and palliative care and targeted interventions to treat pain and disabling symptoms. If our goal is to recognize the full impact of cancer on the lives of patients and their families, it is important to address the emotional and psychological toll of diagnosis and treatment. Recognizing depressive symptoms and clinical depression is an important step toward optimizing the quality of life for patients with cancer.

Data summarized in an excellent review by Pirl published in 2004 show that up to one in five Americans will experience depressive symptoms over the course of their lifetime and that approximately 10%–25% of cancer patients meet criteria for clinical depression.[1] and [2] As our ability to treat depression has improved over the years, thanks in great part to advances in pharmacology and behavioral therapies, it is now critically important to recognize and treat this debilitating disease in individuals with cancer.3 Evidence exists that untreated depression is associated with a worse overall survival for some cancer patients and, paradoxically, that up to half of patients with cancer and concurrent depression are undertreated or receive no treatment.[4], [5] and [6] Medical oncologists receive little or no formal training in psycho-oncology yet are often faced with patients who exhibit changes in mood and become progressively disabled by psychiatric symptoms. Methodical assessment and frequent inquiry may identify patients with cancer and depression.

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

A diagnosis of cancer often precipitates intense emotions such as fear, sadness, and sometimes anger.2 Individuals who may never have given much thought to their own death are confronted with the very real possibility of a shortened life and future suffering. Roles and relationships shift, careers are interrupted, and daily routines may be sacrificed to make room for cancer treatment. Add to this the financial worries that often accompany a serious illness and it is not surprising that patients may require some level of professional guidance or intervention in order to cope with the crisis. As a quick rule of thumb, it takes about 3–4 weeks after diagnosis to adjust, and during that period it is quite normal for patients to experience intense feelings.7 Weissman and Worden, among the first psychiatrists to study distress in cancer patients, described an acute syndrome of distress over existential plight with the diagnosis and with a recurrence that lasts about 100 days.8 Most individuals, given time and adequate support, will find the inner resources to cope with distressing symptoms and find a new normal. Not all do however, and it is important for oncologists to inquire at regular intervals about how the patient is feeling and coping with illness. A recent study by Lo et al9 found that predictors of depressive symptoms in patients with solid tumors included younger age, antidepressant use at baseline, lower self-esteem and spiritual well-being, greater attachment anxiety, hopelessness, the physical burden of symptoms, and proximity to death.

To facilitate screening for emotional distress in the context of a diagnosis of cancer, the National Comprehensive Cancer Network (NCCN) established guidelines that provide a reproducible algorithm for triaging patients with a suspected depression to mental health professionals.10 These guidelines were updated in 2010 and are widely available.11 The consensus definition of distress in cancer is “a multifactorial, unpleasant emotional experience of a psychological (cognitive, behavioral, emotional), social, and/or spiritual nature that may interfere with the ability to cope effectively with cancer, its physical symptoms and its treatment. Distress extends along a continuum, ranging from common feelings of vulnerability, sadness, and fears to problems that can become disabling, such as depression, anxiety, panic, social isolation, and existential and spiritual crisis.”10 By framing distress as a very broad concept, the guidelines separate the broad gamut of normal emotions from the distinct psychiatric syndromes of anxiety and depression which require specialized professional interventions.12

Distress may be a normal response to a threat or crisis, but depressive symptoms should alert the clinician that something more serious is going on. The appearance of persistent symptoms of dysphoria, hopelessness, helplessness, loss of self-esteem, feelings of worthlessness, and suicidal ideation indicates a psychiatric illness.13 The DSM-IV defines a major depressive episode as experiencing either dysphoria or anhedonia in addition to at least five somatic symptoms for at least 2 weeks.14 These somatic symptoms may well overlap with those experienced by patients as a direct result of their cancer or its treatment. Among these are changes in appetite, weight, or sleep; fatigue; loss of energy; and a diminished ability to think or concentrate. The challenge for clinicians is to tease apart the physiologic consequences of disease and side effects of medications from those due to profound and disabling psychiatric syndromes.

Many symptoms caused by cancer itself can be confused with neurovegetative symptoms of depression. Pain is known to modulate the reporting of symptoms; fatigue and weight changes are often secondary to cancer treatment or the illness itself. Patients often feel fatigued due to the heightened metabolic state present when there is a high burden of disease, and cytokines elevated in malignancy have been shown to cause fatigue and appetite suppression. There is a growing literature regarding the development of aberrant sleep patterns in patients with cancer, which can be mistaken for depressive daytime somnolence or insomnia.[15], [16], [17] and [18] Some cancers themselves are associated with a higher risk of depressive symptoms, including pancreatic cancer and cancers of the head and neck.[19], [20] and [21] Chemotherapy can also induce fatigue, insomnia, and anhedonia, as can the steroids often used concomitantly with chemotherapeutic or biologic agents. Interferon-alpha, used to treat melanoma and renal cell cancer, has been associated with depression in 3%–40% of patients; and there is a 5% rate of suicidal thoughts.22

Cancer patients exhibit a range of coping styles and varying degrees of emotional resiliency. If a patient is able to process his or her emotional responses to the physical threat of a diagnosis and becomes mobilized in such a way that he or she obtains useful information and is able to prioritize concerns, obtain social support, and move toward a coherent treatment plan, one can easily assume that he or she is coping well.23 On the other hand, if the patient appears unable to make a decision about treatment, avoids addressing or discussing important issues, and retreats from family, friends, and/or the medical team, one can infer that he or she is having trouble coping and could benefit from a referral to a mental health professional for evaluation.23 Known risk factors for poor coping and for developing depression include social isolation, use of few coping strategies, a history of recent losses or multiple obligations, inflexible coping strategies, the presence of pain, and socioeconomic pressures.[8] and [23] In extreme cases, patients may resort to deferring decisions or simply denying the problem.

Keep in mind there may also be cultural or personal barriers that interfere with a timely and accurate diagnosis of depression.12 Many families believe strongly in the “power of positive thinking” and need to feel that their family member is a “fighter.” This type of encouragement may at times be helpful for a patient, but it may not leave a safe opening for the expression of fear, pain, or depressed mood. If the matriarch or patriarch of the family has supported everyone else through the difficulties in their lives, she or he may not feel able to show weakness and seek help for depression. This can be a difficult patient to diagnose as the only clue to suffering may be easy to miss. In fact, if there are very few questions or complaints when there is clear physical suffering, one needs to worry that the patient is unable to express his or her deep concerns. The clinician who spots this situation early on may be able to lead the patient in the direction of expressing his or her feelings by suggesting that others in similar situations also experience stress or sadness. Finding a private time to talk, away from family members, may also provide a more comfortable environment for a candid conversation.

If we think of the disease trajectory as a marathon, then we can learn to recognize certain landmarks along the course and remember that these pose enormous challenges to patients. In addition to receiving the initial diagnosis, the period of active treatment, the conclusion of active treatment, and the time of disease recurrence pose specific challenges and precipitate intense emotions. Disease recurrence is a time of great anxiety when there is a need to plan for future treatment and an upheaval of the timeline a patient may have made.24

Should the Oncologist Offer Treatment for Depression?

Oncologists assume an important role in the medical care of their patients and often initiate or modify treatments for other medical conditions. If a patient develops hypertension or diabetes during or as a direct consequence of treatment, most oncologists feel comfortable starting medication and may then comanage the patient with internists. Primary care physicians and oncologists are typically familiar with a few basic antidepressants, and many are willing to prescribe these for patients who meet the diagnostic criteria for depression, especially since it takes weeks to achieve adequate therapeutic levels for many of these drugs. Recognizing the presence of depression is thus a key diagnostic intervention.

Several efforts have been made to develop self-report screening inventories that can improve the accuracy and efficiency of detection of depressive symptoms and are brief enough to administer in the setting of an office visit. Some tools have been validated and correlate well with more detailed inventories, although the gold standard remains the detailed psychiatric interview.25 A single-item interview screening proposed by Chochinov et al25 years ago performs as well as or better than longer instruments and is remarkably simple to remember. Asking patients “Are you depressed?” in a brief screening interview correctly identified the eventual diagnostic outcome of every patient in initial studies and has been adopted broadly by oncologists and palliative care clinicians caring for patients who are terminally ill.

We support immediate referral to a psychiatrist for any patient who exhibits symptoms of depression, and there is universal agreement that any person who may be suicidal should be referred immediately for urgent psychiatric evaluation. In practice, however, there are two main barriers to successful referrals for those who may be considered to be “managing” and not considered at risk for suicide: Patients are sometimes resistant to or reluctant to accept a recommendation for referral, and the shortage of mental health professionals trained in psycho-oncology limits quick access. It is, therefore, not surprising that cancer clinicians often initiate pharmacologic therapy for depression and provide emotional support to patients and families. Kadan-Lottick and colleagues5 reported that although 90% of patients agreed that they were willing to receive treatment for emotional distress associated with their cancer diagnosis, only 28% accessed treatment. Approximately 55% of the patients diagnosed in that study with major psychiatric disorders did not access treatment. It has been our experience that oncologists are often willing to initiate pharmacologic therapy while the patient is waiting for an appointment with a specialist.

The most frequently prescribed antidepressant medications are the selective serotonin reuptake inhibitors (SSRIs). Frequently, the choice of antidepressant is based on the side-effect profile of a particular medication as there are many effective options, none of which appears to be significantly more efficacious than the others.7 Antidepressants considered to be sedating may not be the preferred option for patients who have significant neurovegetative symptoms including fatigue and low energy. Conversely, antidepressants that cause anorexia and insomnia are poor options for patients experiencing sleepless nights and continued weight loss. Options for more activating antidepressants include sertraline, escitalopram, bupropion, and venlafaxine, while more sedating antidepressant medications include paroxetine and mirtazapine.7 Methylphenidate, a drug frequently used to treat attention-deficit/hyperactivity disorder, has been very effective in patients with low energy and anorexia.[26] and [27] Starting at a low dose in the morning, especially in the elderly, helps to minimize tachycardia and sleeplessness, which can be unwanted side effects of this medication. Lastly, a key point when choosing a medication is the potential for drug–drug interactions. Multiple antidepressants, including paroxetine, fluoxetine, fluvoxamine, and bupropion, interact with the cytochrome P-450 2D6 system, making them more likely to interact with medications commonly used in oncology.28 One example of this potential for interaction occurs with tamoxifen, which is metabolized into its active form, endoxifen, by the cytochrome P-450 2D6 system. It may not be available in adequate concentrations in the setting of antidepressant medications like paroxetine, an inhibitor of cytochrome P-450 2D6. Whether this ultimately influences the efficacy of anticancer treatment is still under investigation.

While psychotherapy is outside the scope of most practicing oncologists, it may be helpful to provide patients with some guidance about the range of available therapies. Individuals may express a clear preference for nonpharmacologic treatments, so it is important for cancer clinicians to familiarize themselves with a few such options. These include cognitive behavioral therapy (CBT), intensive psychotherapy, and group therapy. These interventions can aid patients in reducing anxiety and in strengthening their personal coping mechanisms. Studies to rigorously evaluate the efficacy of these interventions have been challenging to complete because of the lack of a “gold standard” definition of depression in cancer, no consensus on an appropriate length of treatment, no clear way to monitor compliance with a given therapy, and varied definitions of appropriate end points.12 Despite the challenges, several meta-analyses have been compiled to sort through the data. The more commonly referenced meta-analyses have included thousands of patients undergoing nonpharmacologic interventions ranging from individual psychotherapy to group therapy as far back as 1954.[29], [30], [31], [32], [33] and [34] None of the interventions indicate that any particular therapy is more clearly beneficial than another.

CBT has received recent attention and appears to be a good option for many cancer patients with depression. A review by Williams and Dale in the British Journal of Cancer in 200633 outlines 10 studies focusing on the use of CBT in cancer patients with mixed results. Of these, only two found CBT to be ineffective, whereas the rest demonstrated some benefit in reduction of depressive symptoms and improvement in quality of life for patients with a wide assortment of primary malignancies. Most found early improvement in symptoms but not necessarily long-term persistence of the initial positive effects. Group therapy has also been thoroughly studied in depression in cancer patients since Spiegel's landmark study in the late 1980s and has been shown to decrease anxiety, depression, and pain and to increase effective coping.[34], [35], [36], [37], [38] and [39] Many patients report positive experiences in support groups, but others express an intuitive fear that listening to other patients' concerns and negative thoughts will impair their own overall mood and outlook. Not all patients feel comfortable expressing their personal fears, doubts, and frustrations with a group of relative strangers. Any of these concerns is a sufficient reason to advise more personalized attention in a private therapy session with a specialist. Choosing between individual psychotherapy, group, and family therapy can be construed as another aspect of providing truly “personalized” cancer care.

A substantial number of patients worldwide turn to complementary and alternative therapies for the treatment of cancer and cancer-related symptoms.[40], [41] and [42] Estimates of the prevalence of complementary and alternative therapy use vary widely due to differences in definitions and inaccuracies in self-reporting and patient selection. There are emerging data that up to 60%–80% of cancer patients avail themselves of some form of alternative therapy at some point in the trajectory of their disease.42 This number varies widely, likely because the definition of “complementary and alternative therapies” is so broad and can include prayer, use of herbal medications, acupuncture, and meditation. In one study of early-stage breast cancer patients, the use of alternative medicine was significantly associated with patients experiencing depressive symptoms, heightened fear of recurrence, greater physical symptoms, and poor sexual satisfaction.42 At 1 year, all patients, both those using complementary and alternative therapies and those using traditional methods of care, experienced an improvement in quality of life.

For patients who do not meet the criteria for clinical depression and have no interest in or access to support groups, it is worth remembering there are other interventions that can facilitate adjustment and diminish symptoms of anxiety. Expressive writing, music, or art therapy and other activity-based therapies may provide the necessary vehicles for self-expression.

Conclusion

Depression clearly affects patients with cancer, and establishing the depression diagnosis is the first step toward progress in treatment. Despite the challenges, diagnosis is possible by establishing that the symptoms of depression are negatively impacting patients' abilities to cope with their circumstances and maintain balance in their lives. It is critical not only to make the diagnosis of depression but also to strongly encourage patients to seek treatment, either through pharmacologic or nonpharmacologic means. While we make every effort to eradicate our patients' malignancies, we owe it to them to work just as diligently to improve their daily lives by treating associated depression.

Acknowledgments

We thank Dr. Donna Greenberg and Dr. William Pirl for their thoughtful review of this manuscript and helpful comments.

References2

1 W.F. Pirl, Evidence report on the occurrence, assessment, and treatment of depression in cancer patients, J Natl Cancer Inst Monogr 32 (2004), pp. 32–39. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)

2 J.S. McDaniel, D.L. Musselman, M.R. Porter, D.A. Reed and C.B. Nemeroff, Depression in patients with cancer: diagnosis, biology, and treatment, Arch Gen Psychiatry 52 (1995), pp. 89–99. View Record in Scopus | Cited By in Scopus (339)

3 R.W. Trijsburg, F.C.E. Van Knippenberg and S.E. Rijpma, Effects of psychological treatments on cancer patients: a critical review, Psychosom Med 54 (1992), pp. 489–517. View Record in Scopus | Cited By in Scopus (171)

4 A. Gruneir, T.F. Smith, J. Hirdes and R. Cameron, Depression in patients with advanced illness: an examination of Ontario complex continuing care using the minimum data set 2.0, Palliat Support Care 3 (2005), pp. 99–105. View Record in Scopus | Cited By in Scopus (6)

5 N.S. Kadan-Lottick, L.C. Vanderwerker, S.D. Block, B. Zhang and H.G. Prigerson, Psychiatric disorders and mental health service use in patients with advanced cancer: a report from the Coping with Cancer Study, Cancer 104 (2005), pp. 2872–2881. View Record in Scopus | Cited By in Scopus (57)

6 J.L. Steel, D.A. Geller, T.C. Gamblin, M.C. Olek and B.I. Carr, Depression, immunity, and survival in patients with hepatobiliary carcinoma, J Clin Oncol 25 (2007), pp. 4526–4527.

7 W. Pirl, Depression, anxiety, and fatigue. In: B. Chabner, J. Lynch and D. Longo, Editors, Harrison's Manual of Oncology, McGraw-Hill, New York (2008), pp. 190–196.

8 A. Weissman and J. Worden, The existential plight in cancer: significance of the first 100 days, Psychiatr Med 7 (1976), pp. 1–15.

9 C. Lo, C. Zimmermann and A. Rydall et al., Longitudinal study of depressive symptoms in patients with metastatic gastrointestinal and lung cancer, J Clin Oncol 28 (18) (2010), pp. 3084–3089. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)

10 National Comprehensive Cancer Network, NCCN practice guidelines for the management of psychosocial distress, Oncology (Williston Park) 13 (1999), pp. 113–147.

11 National Comprehensive Cancer Network, NCCN clinical practice guidelines in oncology, Distress management (2010) V.1. www.nccn.org.

12 M. Fisch, Treatment of depression in cancer, J Natl Cancer Inst Monogr 32 (2004), pp. 105–111. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (45)

13 H.T. Mermelstein and L. Lesko, Depression in patients with cancer, Psychooncology 1 (1992), pp. 199–215. Full Text via CrossRef

14 American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders (4th ed.), American Psychiatric Association, Washington DC (1994).

15 M.L. Chen, C.T. Yu and C.H. Yang, Sleep disturbances and quality of life in lung cancer patients undergoing chemotherapy, Lung Cancer 62 (2008), pp. 391–400. Article |

PDF (162 K)
| View Record in Scopus | Cited By in Scopus (8)

16 J. Savard, S. Simard and J. Blanchet et al., Prevalence, clinical characteristics, and risk factors for insomnia in the context of breast cancer, Sleep 24 (2001), pp. 583–590. View Record in Scopus | Cited By in Scopus (81)

17 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)

18 O.G. Palesh, J.A. Roscoe, K.M. Mustian, T. Roth, J. Savard, S. Ancoli-Israel, C. Heckler, J.Q. Purnell, M.C. Janelsins and G.R. Morrow, Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 28 (2010), pp. 292–298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

19 I. Fras, E.M. Litin and J.S. Pearson, Comparison of psychiatric symptoms in carcinoma of the pancreas with those in some other intraabdominal neoplasms, Am J Psychiatry 123 (1967), pp. 1553–1562. View Record in Scopus | Cited By in Scopus (38)

20 R.T. Joffe, D.R. Rubinow, K.D. Denicoff, M. Maher and W.F. Sindelar, Depression and carcinoma of the pancreas, Gen Hosp Psychiatry 8 (1986), pp. 241–245. Article |

PDF (583 K)
| View Record in Scopus | Cited By in Scopus (37)

21 R.P. Morton, A.D.M. Davies, J. Baker, G.A. Baker and P.M. Stell, Quality of life in treated head and neck cancer patients: a preliminary report, Clin Otolaryngol 9 (1984), pp. 181–185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (59)

22 , Micromedex 2.0. DrugPoint summary, interferon alfa-2b http://www.micromedex.com/2 Accessed July 1, 2010.

23 D. Spiegel, A 43-year-old woman coping with cancer, JAMA 281 (4) (1999), pp. 371–377.

24 D.F. Cella, S.M. Mahon and M.I. Donovan, Cancer recurrence as a traumatic event, Behav Med 16 (1990), pp. 15–22. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (126)

25 H.M. Chochinov, K.G. Wilson, M. Enns and S. Lander, “Are you depressed?”: Screening for depression in the terminally ill, Am J Psychiatry 154 (1997), pp. 674–676. View Record in Scopus | Cited By in Scopus (225)

26 E. Bruera, L. Driver and E. Barnes et al., Patient controlled methylphenidate for cancer-related fatigue: a preliminary report, Proc Annu Meet Am Soc Clin Oncol 22 (2003), p. 737.

27 J. Homsi, K.A. Nelson and N. Sarhill et al., A phase II study of methylphenidate for depression in advanced cancer, Am J Hosp Palliat Care 18 (2001), pp. 403–407. View Record in Scopus | Cited By in Scopus (49)

28 G.R. Kalash, Psychotropic drug metabolism in the cancer patient: clinical aspects of management of potential drug interactions, Psychooncology 7 (1998), pp. 307–320. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

29 E.C. Devine and S.K. Westlake, The effects of psychoeducational care provided by adults with cancer: meta-analysis of 116 studies, Oncol Nurs Forum 22 (1995), pp. 1369–1381. View Record in Scopus | Cited By in Scopus (192)

30 T.J. Meyer and M.M. Mark, Effects of psychosocial interventions with adult cancer patients: a meta-analysis of randomized experiments, Health Psychol 14 (1995), pp. 101–108. Abstract |

PDF (3776 K)
| Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (475)

31 S.A. Newell, R.W. Sanson-Fisher and N.J. Savolainen, Systematic review of psychological therapies for cancer patients: overview and recommendations for the future, J Natl Cancer Inst 94 (2002), pp. 558–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (201)

32 T. Sheard and P. Maguire, The effect of psychological interventions on anxiety and depression in cancer patients; results of two meta-analyses, Br J Cancer 80 (1999), pp. 1770–1780. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (210)

33 S. Williams and J. Dale, The effectiveness of treatment for depression/depressive symptoms in adults with cancer: a systematic review, Br J Cancer 94 (2006), pp. 372–390. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (60)

34 D. Spiegel, J.R. Bloom, H.C. Kraemer and E. Gottheil, Effect of psychosocial treatment on survival of patients with metastatic breast cancer, Lancet 2 (1989), pp. 888–891. Article |

PDF (618 K)
| View Record in Scopus | Cited By in Scopus (1113)

35 L.F. Berkman and S.L. Syme, Social networks, host resistence, and mortality: a nine year follow-up study of Alameda County residents, Am J Epidemiol 109 (1979), pp. 186–204. View Record in Scopus | Cited By in Scopus (1297)

36 D.P. Funch and J. Marshall, The role of stress, social support and age in survival from breast cancer, J Psychosom Res 27 (1983), pp. 77–83. Abstract |

PDF (553 K)
| View Record in Scopus | Cited By in Scopus (62)

37 D.C. Ganster and B. Victor, The impact of social support on mental and physical health, Br J Med Psychol 61 (1988), pp. 17–36. View Record in Scopus | Cited By in Scopus (17)

38 F.I. Fawzy, N. Cousins, N.W. Fawzy, M.E. Kemeny, R. Elashoff and D. Morton, A structured psychiatric intervention for cancer patients: I: Changes over time in methods of coping and affective disturbance, Arch Gen Psychiatry 47 (1990), pp. 720–725. View Record in Scopus | Cited By in Scopus (331)

39 D. Spiegel and J.R. Bloom, Group therapy and hypnosis reduce metastatic breast carcinoma pain, Psychosom Med 45 (1983), pp. 333–339. View Record in Scopus | Cited By in Scopus (192)

40 T. Gansler, C. Kaw, C. Crammer and T. Smith, A population-based study of prevalence of complementary methods use by cancer survivors: a report from the American Cancer Society's studies of cancer survivors, Cancer 113 (2008), pp. 1048–1057. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

41 M.A. Richardson, T. Sanders, J.L. Palmer, A. Greisinger and S.E. Singletary, Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology, J Clin Oncol 18 (13) (2000), pp. 2505–2514. View Record in Scopus | Cited By in Scopus (407)

42 H.J. Burstein, S. Gelber, E. Guadagnoli and J.C. Weeks, Use of alternative medicine by women with early-stage breast cancer, N Engl J Med 340 (22) (1999), pp. 1733–1739. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (352)

Conflicts of interest: None to disclose.

Correspondence to: Alicia K. Morgans, MD, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114; telephone: (617) 724-4000; fax: (617) 643-0798


2 PubMed ID in brackets

Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

How we do it

Recognizing Depression in Patients with Cancer

Alicia Morgans MD
,
and Lidia Schapira MD

Received 2 August 2010; 

accepted 20 December 2010. 

Available online 2 April 2011.

Article Outline

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

Should the Oncologist Offer Treatment for Depression?

Conclusion

Acknowledgements

References

Caring for the “whole person” across the continuum of disease remains a top priority for clinicians. Strides in basic and translational research have opened new therapeutic pathways that provide better targeted and effective treatments with fewer side effects. New ancillary and supportive therapies have transformed and improved the experiences of patients undergoing anticancer treatments. Also, with increasing acceptance of multidisciplinary teams, we have the opportunity to make timely referrals to colleagues who provide supportive and palliative care and targeted interventions to treat pain and disabling symptoms. If our goal is to recognize the full impact of cancer on the lives of patients and their families, it is important to address the emotional and psychological toll of diagnosis and treatment. Recognizing depressive symptoms and clinical depression is an important step toward optimizing the quality of life for patients with cancer.

Data summarized in an excellent review by Pirl published in 2004 show that up to one in five Americans will experience depressive symptoms over the course of their lifetime and that approximately 10%–25% of cancer patients meet criteria for clinical depression.[1] and [2] As our ability to treat depression has improved over the years, thanks in great part to advances in pharmacology and behavioral therapies, it is now critically important to recognize and treat this debilitating disease in individuals with cancer.3 Evidence exists that untreated depression is associated with a worse overall survival for some cancer patients and, paradoxically, that up to half of patients with cancer and concurrent depression are undertreated or receive no treatment.[4], [5] and [6] Medical oncologists receive little or no formal training in psycho-oncology yet are often faced with patients who exhibit changes in mood and become progressively disabled by psychiatric symptoms. Methodical assessment and frequent inquiry may identify patients with cancer and depression.

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

A diagnosis of cancer often precipitates intense emotions such as fear, sadness, and sometimes anger.2 Individuals who may never have given much thought to their own death are confronted with the very real possibility of a shortened life and future suffering. Roles and relationships shift, careers are interrupted, and daily routines may be sacrificed to make room for cancer treatment. Add to this the financial worries that often accompany a serious illness and it is not surprising that patients may require some level of professional guidance or intervention in order to cope with the crisis. As a quick rule of thumb, it takes about 3–4 weeks after diagnosis to adjust, and during that period it is quite normal for patients to experience intense feelings.7 Weissman and Worden, among the first psychiatrists to study distress in cancer patients, described an acute syndrome of distress over existential plight with the diagnosis and with a recurrence that lasts about 100 days.8 Most individuals, given time and adequate support, will find the inner resources to cope with distressing symptoms and find a new normal. Not all do however, and it is important for oncologists to inquire at regular intervals about how the patient is feeling and coping with illness. A recent study by Lo et al9 found that predictors of depressive symptoms in patients with solid tumors included younger age, antidepressant use at baseline, lower self-esteem and spiritual well-being, greater attachment anxiety, hopelessness, the physical burden of symptoms, and proximity to death.

To facilitate screening for emotional distress in the context of a diagnosis of cancer, the National Comprehensive Cancer Network (NCCN) established guidelines that provide a reproducible algorithm for triaging patients with a suspected depression to mental health professionals.10 These guidelines were updated in 2010 and are widely available.11 The consensus definition of distress in cancer is “a multifactorial, unpleasant emotional experience of a psychological (cognitive, behavioral, emotional), social, and/or spiritual nature that may interfere with the ability to cope effectively with cancer, its physical symptoms and its treatment. Distress extends along a continuum, ranging from common feelings of vulnerability, sadness, and fears to problems that can become disabling, such as depression, anxiety, panic, social isolation, and existential and spiritual crisis.”10 By framing distress as a very broad concept, the guidelines separate the broad gamut of normal emotions from the distinct psychiatric syndromes of anxiety and depression which require specialized professional interventions.12

Distress may be a normal response to a threat or crisis, but depressive symptoms should alert the clinician that something more serious is going on. The appearance of persistent symptoms of dysphoria, hopelessness, helplessness, loss of self-esteem, feelings of worthlessness, and suicidal ideation indicates a psychiatric illness.13 The DSM-IV defines a major depressive episode as experiencing either dysphoria or anhedonia in addition to at least five somatic symptoms for at least 2 weeks.14 These somatic symptoms may well overlap with those experienced by patients as a direct result of their cancer or its treatment. Among these are changes in appetite, weight, or sleep; fatigue; loss of energy; and a diminished ability to think or concentrate. The challenge for clinicians is to tease apart the physiologic consequences of disease and side effects of medications from those due to profound and disabling psychiatric syndromes.

Many symptoms caused by cancer itself can be confused with neurovegetative symptoms of depression. Pain is known to modulate the reporting of symptoms; fatigue and weight changes are often secondary to cancer treatment or the illness itself. Patients often feel fatigued due to the heightened metabolic state present when there is a high burden of disease, and cytokines elevated in malignancy have been shown to cause fatigue and appetite suppression. There is a growing literature regarding the development of aberrant sleep patterns in patients with cancer, which can be mistaken for depressive daytime somnolence or insomnia.[15], [16], [17] and [18] Some cancers themselves are associated with a higher risk of depressive symptoms, including pancreatic cancer and cancers of the head and neck.[19], [20] and [21] Chemotherapy can also induce fatigue, insomnia, and anhedonia, as can the steroids often used concomitantly with chemotherapeutic or biologic agents. Interferon-alpha, used to treat melanoma and renal cell cancer, has been associated with depression in 3%–40% of patients; and there is a 5% rate of suicidal thoughts.22

Cancer patients exhibit a range of coping styles and varying degrees of emotional resiliency. If a patient is able to process his or her emotional responses to the physical threat of a diagnosis and becomes mobilized in such a way that he or she obtains useful information and is able to prioritize concerns, obtain social support, and move toward a coherent treatment plan, one can easily assume that he or she is coping well.23 On the other hand, if the patient appears unable to make a decision about treatment, avoids addressing or discussing important issues, and retreats from family, friends, and/or the medical team, one can infer that he or she is having trouble coping and could benefit from a referral to a mental health professional for evaluation.23 Known risk factors for poor coping and for developing depression include social isolation, use of few coping strategies, a history of recent losses or multiple obligations, inflexible coping strategies, the presence of pain, and socioeconomic pressures.[8] and [23] In extreme cases, patients may resort to deferring decisions or simply denying the problem.

Keep in mind there may also be cultural or personal barriers that interfere with a timely and accurate diagnosis of depression.12 Many families believe strongly in the “power of positive thinking” and need to feel that their family member is a “fighter.” This type of encouragement may at times be helpful for a patient, but it may not leave a safe opening for the expression of fear, pain, or depressed mood. If the matriarch or patriarch of the family has supported everyone else through the difficulties in their lives, she or he may not feel able to show weakness and seek help for depression. This can be a difficult patient to diagnose as the only clue to suffering may be easy to miss. In fact, if there are very few questions or complaints when there is clear physical suffering, one needs to worry that the patient is unable to express his or her deep concerns. The clinician who spots this situation early on may be able to lead the patient in the direction of expressing his or her feelings by suggesting that others in similar situations also experience stress or sadness. Finding a private time to talk, away from family members, may also provide a more comfortable environment for a candid conversation.

If we think of the disease trajectory as a marathon, then we can learn to recognize certain landmarks along the course and remember that these pose enormous challenges to patients. In addition to receiving the initial diagnosis, the period of active treatment, the conclusion of active treatment, and the time of disease recurrence pose specific challenges and precipitate intense emotions. Disease recurrence is a time of great anxiety when there is a need to plan for future treatment and an upheaval of the timeline a patient may have made.24

Should the Oncologist Offer Treatment for Depression?

Oncologists assume an important role in the medical care of their patients and often initiate or modify treatments for other medical conditions. If a patient develops hypertension or diabetes during or as a direct consequence of treatment, most oncologists feel comfortable starting medication and may then comanage the patient with internists. Primary care physicians and oncologists are typically familiar with a few basic antidepressants, and many are willing to prescribe these for patients who meet the diagnostic criteria for depression, especially since it takes weeks to achieve adequate therapeutic levels for many of these drugs. Recognizing the presence of depression is thus a key diagnostic intervention.

Several efforts have been made to develop self-report screening inventories that can improve the accuracy and efficiency of detection of depressive symptoms and are brief enough to administer in the setting of an office visit. Some tools have been validated and correlate well with more detailed inventories, although the gold standard remains the detailed psychiatric interview.25 A single-item interview screening proposed by Chochinov et al25 years ago performs as well as or better than longer instruments and is remarkably simple to remember. Asking patients “Are you depressed?” in a brief screening interview correctly identified the eventual diagnostic outcome of every patient in initial studies and has been adopted broadly by oncologists and palliative care clinicians caring for patients who are terminally ill.

We support immediate referral to a psychiatrist for any patient who exhibits symptoms of depression, and there is universal agreement that any person who may be suicidal should be referred immediately for urgent psychiatric evaluation. In practice, however, there are two main barriers to successful referrals for those who may be considered to be “managing” and not considered at risk for suicide: Patients are sometimes resistant to or reluctant to accept a recommendation for referral, and the shortage of mental health professionals trained in psycho-oncology limits quick access. It is, therefore, not surprising that cancer clinicians often initiate pharmacologic therapy for depression and provide emotional support to patients and families. Kadan-Lottick and colleagues5 reported that although 90% of patients agreed that they were willing to receive treatment for emotional distress associated with their cancer diagnosis, only 28% accessed treatment. Approximately 55% of the patients diagnosed in that study with major psychiatric disorders did not access treatment. It has been our experience that oncologists are often willing to initiate pharmacologic therapy while the patient is waiting for an appointment with a specialist.

The most frequently prescribed antidepressant medications are the selective serotonin reuptake inhibitors (SSRIs). Frequently, the choice of antidepressant is based on the side-effect profile of a particular medication as there are many effective options, none of which appears to be significantly more efficacious than the others.7 Antidepressants considered to be sedating may not be the preferred option for patients who have significant neurovegetative symptoms including fatigue and low energy. Conversely, antidepressants that cause anorexia and insomnia are poor options for patients experiencing sleepless nights and continued weight loss. Options for more activating antidepressants include sertraline, escitalopram, bupropion, and venlafaxine, while more sedating antidepressant medications include paroxetine and mirtazapine.7 Methylphenidate, a drug frequently used to treat attention-deficit/hyperactivity disorder, has been very effective in patients with low energy and anorexia.[26] and [27] Starting at a low dose in the morning, especially in the elderly, helps to minimize tachycardia and sleeplessness, which can be unwanted side effects of this medication. Lastly, a key point when choosing a medication is the potential for drug–drug interactions. Multiple antidepressants, including paroxetine, fluoxetine, fluvoxamine, and bupropion, interact with the cytochrome P-450 2D6 system, making them more likely to interact with medications commonly used in oncology.28 One example of this potential for interaction occurs with tamoxifen, which is metabolized into its active form, endoxifen, by the cytochrome P-450 2D6 system. It may not be available in adequate concentrations in the setting of antidepressant medications like paroxetine, an inhibitor of cytochrome P-450 2D6. Whether this ultimately influences the efficacy of anticancer treatment is still under investigation.

While psychotherapy is outside the scope of most practicing oncologists, it may be helpful to provide patients with some guidance about the range of available therapies. Individuals may express a clear preference for nonpharmacologic treatments, so it is important for cancer clinicians to familiarize themselves with a few such options. These include cognitive behavioral therapy (CBT), intensive psychotherapy, and group therapy. These interventions can aid patients in reducing anxiety and in strengthening their personal coping mechanisms. Studies to rigorously evaluate the efficacy of these interventions have been challenging to complete because of the lack of a “gold standard” definition of depression in cancer, no consensus on an appropriate length of treatment, no clear way to monitor compliance with a given therapy, and varied definitions of appropriate end points.12 Despite the challenges, several meta-analyses have been compiled to sort through the data. The more commonly referenced meta-analyses have included thousands of patients undergoing nonpharmacologic interventions ranging from individual psychotherapy to group therapy as far back as 1954.[29], [30], [31], [32], [33] and [34] None of the interventions indicate that any particular therapy is more clearly beneficial than another.

CBT has received recent attention and appears to be a good option for many cancer patients with depression. A review by Williams and Dale in the British Journal of Cancer in 200633 outlines 10 studies focusing on the use of CBT in cancer patients with mixed results. Of these, only two found CBT to be ineffective, whereas the rest demonstrated some benefit in reduction of depressive symptoms and improvement in quality of life for patients with a wide assortment of primary malignancies. Most found early improvement in symptoms but not necessarily long-term persistence of the initial positive effects. Group therapy has also been thoroughly studied in depression in cancer patients since Spiegel's landmark study in the late 1980s and has been shown to decrease anxiety, depression, and pain and to increase effective coping.[34], [35], [36], [37], [38] and [39] Many patients report positive experiences in support groups, but others express an intuitive fear that listening to other patients' concerns and negative thoughts will impair their own overall mood and outlook. Not all patients feel comfortable expressing their personal fears, doubts, and frustrations with a group of relative strangers. Any of these concerns is a sufficient reason to advise more personalized attention in a private therapy session with a specialist. Choosing between individual psychotherapy, group, and family therapy can be construed as another aspect of providing truly “personalized” cancer care.

A substantial number of patients worldwide turn to complementary and alternative therapies for the treatment of cancer and cancer-related symptoms.[40], [41] and [42] Estimates of the prevalence of complementary and alternative therapy use vary widely due to differences in definitions and inaccuracies in self-reporting and patient selection. There are emerging data that up to 60%–80% of cancer patients avail themselves of some form of alternative therapy at some point in the trajectory of their disease.42 This number varies widely, likely because the definition of “complementary and alternative therapies” is so broad and can include prayer, use of herbal medications, acupuncture, and meditation. In one study of early-stage breast cancer patients, the use of alternative medicine was significantly associated with patients experiencing depressive symptoms, heightened fear of recurrence, greater physical symptoms, and poor sexual satisfaction.42 At 1 year, all patients, both those using complementary and alternative therapies and those using traditional methods of care, experienced an improvement in quality of life.

For patients who do not meet the criteria for clinical depression and have no interest in or access to support groups, it is worth remembering there are other interventions that can facilitate adjustment and diminish symptoms of anxiety. Expressive writing, music, or art therapy and other activity-based therapies may provide the necessary vehicles for self-expression.

Conclusion

Depression clearly affects patients with cancer, and establishing the depression diagnosis is the first step toward progress in treatment. Despite the challenges, diagnosis is possible by establishing that the symptoms of depression are negatively impacting patients' abilities to cope with their circumstances and maintain balance in their lives. It is critical not only to make the diagnosis of depression but also to strongly encourage patients to seek treatment, either through pharmacologic or nonpharmacologic means. While we make every effort to eradicate our patients' malignancies, we owe it to them to work just as diligently to improve their daily lives by treating associated depression.

Acknowledgments

We thank Dr. Donna Greenberg and Dr. William Pirl for their thoughtful review of this manuscript and helpful comments.

References2

1 W.F. Pirl, Evidence report on the occurrence, assessment, and treatment of depression in cancer patients, J Natl Cancer Inst Monogr 32 (2004), pp. 32–39. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)

2 J.S. McDaniel, D.L. Musselman, M.R. Porter, D.A. Reed and C.B. Nemeroff, Depression in patients with cancer: diagnosis, biology, and treatment, Arch Gen Psychiatry 52 (1995), pp. 89–99. View Record in Scopus | Cited By in Scopus (339)

3 R.W. Trijsburg, F.C.E. Van Knippenberg and S.E. Rijpma, Effects of psychological treatments on cancer patients: a critical review, Psychosom Med 54 (1992), pp. 489–517. View Record in Scopus | Cited By in Scopus (171)

4 A. Gruneir, T.F. Smith, J. Hirdes and R. Cameron, Depression in patients with advanced illness: an examination of Ontario complex continuing care using the minimum data set 2.0, Palliat Support Care 3 (2005), pp. 99–105. View Record in Scopus | Cited By in Scopus (6)

5 N.S. Kadan-Lottick, L.C. Vanderwerker, S.D. Block, B. Zhang and H.G. Prigerson, Psychiatric disorders and mental health service use in patients with advanced cancer: a report from the Coping with Cancer Study, Cancer 104 (2005), pp. 2872–2881. View Record in Scopus | Cited By in Scopus (57)

6 J.L. Steel, D.A. Geller, T.C. Gamblin, M.C. Olek and B.I. Carr, Depression, immunity, and survival in patients with hepatobiliary carcinoma, J Clin Oncol 25 (2007), pp. 4526–4527.

7 W. Pirl, Depression, anxiety, and fatigue. In: B. Chabner, J. Lynch and D. Longo, Editors, Harrison's Manual of Oncology, McGraw-Hill, New York (2008), pp. 190–196.

8 A. Weissman and J. Worden, The existential plight in cancer: significance of the first 100 days, Psychiatr Med 7 (1976), pp. 1–15.

9 C. Lo, C. Zimmermann and A. Rydall et al., Longitudinal study of depressive symptoms in patients with metastatic gastrointestinal and lung cancer, J Clin Oncol 28 (18) (2010), pp. 3084–3089. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)

10 National Comprehensive Cancer Network, NCCN practice guidelines for the management of psychosocial distress, Oncology (Williston Park) 13 (1999), pp. 113–147.

11 National Comprehensive Cancer Network, NCCN clinical practice guidelines in oncology, Distress management (2010) V.1. www.nccn.org.

12 M. Fisch, Treatment of depression in cancer, J Natl Cancer Inst Monogr 32 (2004), pp. 105–111. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (45)

13 H.T. Mermelstein and L. Lesko, Depression in patients with cancer, Psychooncology 1 (1992), pp. 199–215. Full Text via CrossRef

14 American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders (4th ed.), American Psychiatric Association, Washington DC (1994).

15 M.L. Chen, C.T. Yu and C.H. Yang, Sleep disturbances and quality of life in lung cancer patients undergoing chemotherapy, Lung Cancer 62 (2008), pp. 391–400. Article |

PDF (162 K)
| View Record in Scopus | Cited By in Scopus (8)

16 J. Savard, S. Simard and J. Blanchet et al., Prevalence, clinical characteristics, and risk factors for insomnia in the context of breast cancer, Sleep 24 (2001), pp. 583–590. View Record in Scopus | Cited By in Scopus (81)

17 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)

18 O.G. Palesh, J.A. Roscoe, K.M. Mustian, T. Roth, J. Savard, S. Ancoli-Israel, C. Heckler, J.Q. Purnell, M.C. Janelsins and G.R. Morrow, Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 28 (2010), pp. 292–298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

19 I. Fras, E.M. Litin and J.S. Pearson, Comparison of psychiatric symptoms in carcinoma of the pancreas with those in some other intraabdominal neoplasms, Am J Psychiatry 123 (1967), pp. 1553–1562. View Record in Scopus | Cited By in Scopus (38)

20 R.T. Joffe, D.R. Rubinow, K.D. Denicoff, M. Maher and W.F. Sindelar, Depression and carcinoma of the pancreas, Gen Hosp Psychiatry 8 (1986), pp. 241–245. Article |

PDF (583 K)
| View Record in Scopus | Cited By in Scopus (37)

21 R.P. Morton, A.D.M. Davies, J. Baker, G.A. Baker and P.M. Stell, Quality of life in treated head and neck cancer patients: a preliminary report, Clin Otolaryngol 9 (1984), pp. 181–185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (59)

22 , Micromedex 2.0. DrugPoint summary, interferon alfa-2b http://www.micromedex.com/2 Accessed July 1, 2010.

23 D. Spiegel, A 43-year-old woman coping with cancer, JAMA 281 (4) (1999), pp. 371–377.

24 D.F. Cella, S.M. Mahon and M.I. Donovan, Cancer recurrence as a traumatic event, Behav Med 16 (1990), pp. 15–22. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (126)

25 H.M. Chochinov, K.G. Wilson, M. Enns and S. Lander, “Are you depressed?”: Screening for depression in the terminally ill, Am J Psychiatry 154 (1997), pp. 674–676. View Record in Scopus | Cited By in Scopus (225)

26 E. Bruera, L. Driver and E. Barnes et al., Patient controlled methylphenidate for cancer-related fatigue: a preliminary report, Proc Annu Meet Am Soc Clin Oncol 22 (2003), p. 737.

27 J. Homsi, K.A. Nelson and N. Sarhill et al., A phase II study of methylphenidate for depression in advanced cancer, Am J Hosp Palliat Care 18 (2001), pp. 403–407. View Record in Scopus | Cited By in Scopus (49)

28 G.R. Kalash, Psychotropic drug metabolism in the cancer patient: clinical aspects of management of potential drug interactions, Psychooncology 7 (1998), pp. 307–320. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

29 E.C. Devine and S.K. Westlake, The effects of psychoeducational care provided by adults with cancer: meta-analysis of 116 studies, Oncol Nurs Forum 22 (1995), pp. 1369–1381. View Record in Scopus | Cited By in Scopus (192)

30 T.J. Meyer and M.M. Mark, Effects of psychosocial interventions with adult cancer patients: a meta-analysis of randomized experiments, Health Psychol 14 (1995), pp. 101–108. Abstract |

PDF (3776 K)
| Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (475)

31 S.A. Newell, R.W. Sanson-Fisher and N.J. Savolainen, Systematic review of psychological therapies for cancer patients: overview and recommendations for the future, J Natl Cancer Inst 94 (2002), pp. 558–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (201)

32 T. Sheard and P. Maguire, The effect of psychological interventions on anxiety and depression in cancer patients; results of two meta-analyses, Br J Cancer 80 (1999), pp. 1770–1780. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (210)

33 S. Williams and J. Dale, The effectiveness of treatment for depression/depressive symptoms in adults with cancer: a systematic review, Br J Cancer 94 (2006), pp. 372–390. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (60)

34 D. Spiegel, J.R. Bloom, H.C. Kraemer and E. Gottheil, Effect of psychosocial treatment on survival of patients with metastatic breast cancer, Lancet 2 (1989), pp. 888–891. Article |

PDF (618 K)
| View Record in Scopus | Cited By in Scopus (1113)

35 L.F. Berkman and S.L. Syme, Social networks, host resistence, and mortality: a nine year follow-up study of Alameda County residents, Am J Epidemiol 109 (1979), pp. 186–204. View Record in Scopus | Cited By in Scopus (1297)

36 D.P. Funch and J. Marshall, The role of stress, social support and age in survival from breast cancer, J Psychosom Res 27 (1983), pp. 77–83. Abstract |

PDF (553 K)
| View Record in Scopus | Cited By in Scopus (62)

37 D.C. Ganster and B. Victor, The impact of social support on mental and physical health, Br J Med Psychol 61 (1988), pp. 17–36. View Record in Scopus | Cited By in Scopus (17)

38 F.I. Fawzy, N. Cousins, N.W. Fawzy, M.E. Kemeny, R. Elashoff and D. Morton, A structured psychiatric intervention for cancer patients: I: Changes over time in methods of coping and affective disturbance, Arch Gen Psychiatry 47 (1990), pp. 720–725. View Record in Scopus | Cited By in Scopus (331)

39 D. Spiegel and J.R. Bloom, Group therapy and hypnosis reduce metastatic breast carcinoma pain, Psychosom Med 45 (1983), pp. 333–339. View Record in Scopus | Cited By in Scopus (192)

40 T. Gansler, C. Kaw, C. Crammer and T. Smith, A population-based study of prevalence of complementary methods use by cancer survivors: a report from the American Cancer Society's studies of cancer survivors, Cancer 113 (2008), pp. 1048–1057. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

41 M.A. Richardson, T. Sanders, J.L. Palmer, A. Greisinger and S.E. Singletary, Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology, J Clin Oncol 18 (13) (2000), pp. 2505–2514. View Record in Scopus | Cited By in Scopus (407)

42 H.J. Burstein, S. Gelber, E. Guadagnoli and J.C. Weeks, Use of alternative medicine by women with early-stage breast cancer, N Engl J Med 340 (22) (1999), pp. 1733–1739. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (352)

Conflicts of interest: None to disclose.

Correspondence to: Alicia K. Morgans, MD, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114; telephone: (617) 724-4000; fax: (617) 643-0798


2 PubMed ID in brackets

How we do it

Recognizing Depression in Patients with Cancer

Alicia Morgans MD
,
and Lidia Schapira MD

Received 2 August 2010; 

accepted 20 December 2010. 

Available online 2 April 2011.

Article Outline

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

Should the Oncologist Offer Treatment for Depression?

Conclusion

Acknowledgements

References

Caring for the “whole person” across the continuum of disease remains a top priority for clinicians. Strides in basic and translational research have opened new therapeutic pathways that provide better targeted and effective treatments with fewer side effects. New ancillary and supportive therapies have transformed and improved the experiences of patients undergoing anticancer treatments. Also, with increasing acceptance of multidisciplinary teams, we have the opportunity to make timely referrals to colleagues who provide supportive and palliative care and targeted interventions to treat pain and disabling symptoms. If our goal is to recognize the full impact of cancer on the lives of patients and their families, it is important to address the emotional and psychological toll of diagnosis and treatment. Recognizing depressive symptoms and clinical depression is an important step toward optimizing the quality of life for patients with cancer.

Data summarized in an excellent review by Pirl published in 2004 show that up to one in five Americans will experience depressive symptoms over the course of their lifetime and that approximately 10%–25% of cancer patients meet criteria for clinical depression.[1] and [2] As our ability to treat depression has improved over the years, thanks in great part to advances in pharmacology and behavioral therapies, it is now critically important to recognize and treat this debilitating disease in individuals with cancer.3 Evidence exists that untreated depression is associated with a worse overall survival for some cancer patients and, paradoxically, that up to half of patients with cancer and concurrent depression are undertreated or receive no treatment.[4], [5] and [6] Medical oncologists receive little or no formal training in psycho-oncology yet are often faced with patients who exhibit changes in mood and become progressively disabled by psychiatric symptoms. Methodical assessment and frequent inquiry may identify patients with cancer and depression.

Peeling Back the Onion: Sorting through Symptoms to Reach a Diagnosis

A diagnosis of cancer often precipitates intense emotions such as fear, sadness, and sometimes anger.2 Individuals who may never have given much thought to their own death are confronted with the very real possibility of a shortened life and future suffering. Roles and relationships shift, careers are interrupted, and daily routines may be sacrificed to make room for cancer treatment. Add to this the financial worries that often accompany a serious illness and it is not surprising that patients may require some level of professional guidance or intervention in order to cope with the crisis. As a quick rule of thumb, it takes about 3–4 weeks after diagnosis to adjust, and during that period it is quite normal for patients to experience intense feelings.7 Weissman and Worden, among the first psychiatrists to study distress in cancer patients, described an acute syndrome of distress over existential plight with the diagnosis and with a recurrence that lasts about 100 days.8 Most individuals, given time and adequate support, will find the inner resources to cope with distressing symptoms and find a new normal. Not all do however, and it is important for oncologists to inquire at regular intervals about how the patient is feeling and coping with illness. A recent study by Lo et al9 found that predictors of depressive symptoms in patients with solid tumors included younger age, antidepressant use at baseline, lower self-esteem and spiritual well-being, greater attachment anxiety, hopelessness, the physical burden of symptoms, and proximity to death.

To facilitate screening for emotional distress in the context of a diagnosis of cancer, the National Comprehensive Cancer Network (NCCN) established guidelines that provide a reproducible algorithm for triaging patients with a suspected depression to mental health professionals.10 These guidelines were updated in 2010 and are widely available.11 The consensus definition of distress in cancer is “a multifactorial, unpleasant emotional experience of a psychological (cognitive, behavioral, emotional), social, and/or spiritual nature that may interfere with the ability to cope effectively with cancer, its physical symptoms and its treatment. Distress extends along a continuum, ranging from common feelings of vulnerability, sadness, and fears to problems that can become disabling, such as depression, anxiety, panic, social isolation, and existential and spiritual crisis.”10 By framing distress as a very broad concept, the guidelines separate the broad gamut of normal emotions from the distinct psychiatric syndromes of anxiety and depression which require specialized professional interventions.12

Distress may be a normal response to a threat or crisis, but depressive symptoms should alert the clinician that something more serious is going on. The appearance of persistent symptoms of dysphoria, hopelessness, helplessness, loss of self-esteem, feelings of worthlessness, and suicidal ideation indicates a psychiatric illness.13 The DSM-IV defines a major depressive episode as experiencing either dysphoria or anhedonia in addition to at least five somatic symptoms for at least 2 weeks.14 These somatic symptoms may well overlap with those experienced by patients as a direct result of their cancer or its treatment. Among these are changes in appetite, weight, or sleep; fatigue; loss of energy; and a diminished ability to think or concentrate. The challenge for clinicians is to tease apart the physiologic consequences of disease and side effects of medications from those due to profound and disabling psychiatric syndromes.

Many symptoms caused by cancer itself can be confused with neurovegetative symptoms of depression. Pain is known to modulate the reporting of symptoms; fatigue and weight changes are often secondary to cancer treatment or the illness itself. Patients often feel fatigued due to the heightened metabolic state present when there is a high burden of disease, and cytokines elevated in malignancy have been shown to cause fatigue and appetite suppression. There is a growing literature regarding the development of aberrant sleep patterns in patients with cancer, which can be mistaken for depressive daytime somnolence or insomnia.[15], [16], [17] and [18] Some cancers themselves are associated with a higher risk of depressive symptoms, including pancreatic cancer and cancers of the head and neck.[19], [20] and [21] Chemotherapy can also induce fatigue, insomnia, and anhedonia, as can the steroids often used concomitantly with chemotherapeutic or biologic agents. Interferon-alpha, used to treat melanoma and renal cell cancer, has been associated with depression in 3%–40% of patients; and there is a 5% rate of suicidal thoughts.22

Cancer patients exhibit a range of coping styles and varying degrees of emotional resiliency. If a patient is able to process his or her emotional responses to the physical threat of a diagnosis and becomes mobilized in such a way that he or she obtains useful information and is able to prioritize concerns, obtain social support, and move toward a coherent treatment plan, one can easily assume that he or she is coping well.23 On the other hand, if the patient appears unable to make a decision about treatment, avoids addressing or discussing important issues, and retreats from family, friends, and/or the medical team, one can infer that he or she is having trouble coping and could benefit from a referral to a mental health professional for evaluation.23 Known risk factors for poor coping and for developing depression include social isolation, use of few coping strategies, a history of recent losses or multiple obligations, inflexible coping strategies, the presence of pain, and socioeconomic pressures.[8] and [23] In extreme cases, patients may resort to deferring decisions or simply denying the problem.

Keep in mind there may also be cultural or personal barriers that interfere with a timely and accurate diagnosis of depression.12 Many families believe strongly in the “power of positive thinking” and need to feel that their family member is a “fighter.” This type of encouragement may at times be helpful for a patient, but it may not leave a safe opening for the expression of fear, pain, or depressed mood. If the matriarch or patriarch of the family has supported everyone else through the difficulties in their lives, she or he may not feel able to show weakness and seek help for depression. This can be a difficult patient to diagnose as the only clue to suffering may be easy to miss. In fact, if there are very few questions or complaints when there is clear physical suffering, one needs to worry that the patient is unable to express his or her deep concerns. The clinician who spots this situation early on may be able to lead the patient in the direction of expressing his or her feelings by suggesting that others in similar situations also experience stress or sadness. Finding a private time to talk, away from family members, may also provide a more comfortable environment for a candid conversation.

If we think of the disease trajectory as a marathon, then we can learn to recognize certain landmarks along the course and remember that these pose enormous challenges to patients. In addition to receiving the initial diagnosis, the period of active treatment, the conclusion of active treatment, and the time of disease recurrence pose specific challenges and precipitate intense emotions. Disease recurrence is a time of great anxiety when there is a need to plan for future treatment and an upheaval of the timeline a patient may have made.24

Should the Oncologist Offer Treatment for Depression?

Oncologists assume an important role in the medical care of their patients and often initiate or modify treatments for other medical conditions. If a patient develops hypertension or diabetes during or as a direct consequence of treatment, most oncologists feel comfortable starting medication and may then comanage the patient with internists. Primary care physicians and oncologists are typically familiar with a few basic antidepressants, and many are willing to prescribe these for patients who meet the diagnostic criteria for depression, especially since it takes weeks to achieve adequate therapeutic levels for many of these drugs. Recognizing the presence of depression is thus a key diagnostic intervention.

Several efforts have been made to develop self-report screening inventories that can improve the accuracy and efficiency of detection of depressive symptoms and are brief enough to administer in the setting of an office visit. Some tools have been validated and correlate well with more detailed inventories, although the gold standard remains the detailed psychiatric interview.25 A single-item interview screening proposed by Chochinov et al25 years ago performs as well as or better than longer instruments and is remarkably simple to remember. Asking patients “Are you depressed?” in a brief screening interview correctly identified the eventual diagnostic outcome of every patient in initial studies and has been adopted broadly by oncologists and palliative care clinicians caring for patients who are terminally ill.

We support immediate referral to a psychiatrist for any patient who exhibits symptoms of depression, and there is universal agreement that any person who may be suicidal should be referred immediately for urgent psychiatric evaluation. In practice, however, there are two main barriers to successful referrals for those who may be considered to be “managing” and not considered at risk for suicide: Patients are sometimes resistant to or reluctant to accept a recommendation for referral, and the shortage of mental health professionals trained in psycho-oncology limits quick access. It is, therefore, not surprising that cancer clinicians often initiate pharmacologic therapy for depression and provide emotional support to patients and families. Kadan-Lottick and colleagues5 reported that although 90% of patients agreed that they were willing to receive treatment for emotional distress associated with their cancer diagnosis, only 28% accessed treatment. Approximately 55% of the patients diagnosed in that study with major psychiatric disorders did not access treatment. It has been our experience that oncologists are often willing to initiate pharmacologic therapy while the patient is waiting for an appointment with a specialist.

The most frequently prescribed antidepressant medications are the selective serotonin reuptake inhibitors (SSRIs). Frequently, the choice of antidepressant is based on the side-effect profile of a particular medication as there are many effective options, none of which appears to be significantly more efficacious than the others.7 Antidepressants considered to be sedating may not be the preferred option for patients who have significant neurovegetative symptoms including fatigue and low energy. Conversely, antidepressants that cause anorexia and insomnia are poor options for patients experiencing sleepless nights and continued weight loss. Options for more activating antidepressants include sertraline, escitalopram, bupropion, and venlafaxine, while more sedating antidepressant medications include paroxetine and mirtazapine.7 Methylphenidate, a drug frequently used to treat attention-deficit/hyperactivity disorder, has been very effective in patients with low energy and anorexia.[26] and [27] Starting at a low dose in the morning, especially in the elderly, helps to minimize tachycardia and sleeplessness, which can be unwanted side effects of this medication. Lastly, a key point when choosing a medication is the potential for drug–drug interactions. Multiple antidepressants, including paroxetine, fluoxetine, fluvoxamine, and bupropion, interact with the cytochrome P-450 2D6 system, making them more likely to interact with medications commonly used in oncology.28 One example of this potential for interaction occurs with tamoxifen, which is metabolized into its active form, endoxifen, by the cytochrome P-450 2D6 system. It may not be available in adequate concentrations in the setting of antidepressant medications like paroxetine, an inhibitor of cytochrome P-450 2D6. Whether this ultimately influences the efficacy of anticancer treatment is still under investigation.

While psychotherapy is outside the scope of most practicing oncologists, it may be helpful to provide patients with some guidance about the range of available therapies. Individuals may express a clear preference for nonpharmacologic treatments, so it is important for cancer clinicians to familiarize themselves with a few such options. These include cognitive behavioral therapy (CBT), intensive psychotherapy, and group therapy. These interventions can aid patients in reducing anxiety and in strengthening their personal coping mechanisms. Studies to rigorously evaluate the efficacy of these interventions have been challenging to complete because of the lack of a “gold standard” definition of depression in cancer, no consensus on an appropriate length of treatment, no clear way to monitor compliance with a given therapy, and varied definitions of appropriate end points.12 Despite the challenges, several meta-analyses have been compiled to sort through the data. The more commonly referenced meta-analyses have included thousands of patients undergoing nonpharmacologic interventions ranging from individual psychotherapy to group therapy as far back as 1954.[29], [30], [31], [32], [33] and [34] None of the interventions indicate that any particular therapy is more clearly beneficial than another.

CBT has received recent attention and appears to be a good option for many cancer patients with depression. A review by Williams and Dale in the British Journal of Cancer in 200633 outlines 10 studies focusing on the use of CBT in cancer patients with mixed results. Of these, only two found CBT to be ineffective, whereas the rest demonstrated some benefit in reduction of depressive symptoms and improvement in quality of life for patients with a wide assortment of primary malignancies. Most found early improvement in symptoms but not necessarily long-term persistence of the initial positive effects. Group therapy has also been thoroughly studied in depression in cancer patients since Spiegel's landmark study in the late 1980s and has been shown to decrease anxiety, depression, and pain and to increase effective coping.[34], [35], [36], [37], [38] and [39] Many patients report positive experiences in support groups, but others express an intuitive fear that listening to other patients' concerns and negative thoughts will impair their own overall mood and outlook. Not all patients feel comfortable expressing their personal fears, doubts, and frustrations with a group of relative strangers. Any of these concerns is a sufficient reason to advise more personalized attention in a private therapy session with a specialist. Choosing between individual psychotherapy, group, and family therapy can be construed as another aspect of providing truly “personalized” cancer care.

A substantial number of patients worldwide turn to complementary and alternative therapies for the treatment of cancer and cancer-related symptoms.[40], [41] and [42] Estimates of the prevalence of complementary and alternative therapy use vary widely due to differences in definitions and inaccuracies in self-reporting and patient selection. There are emerging data that up to 60%–80% of cancer patients avail themselves of some form of alternative therapy at some point in the trajectory of their disease.42 This number varies widely, likely because the definition of “complementary and alternative therapies” is so broad and can include prayer, use of herbal medications, acupuncture, and meditation. In one study of early-stage breast cancer patients, the use of alternative medicine was significantly associated with patients experiencing depressive symptoms, heightened fear of recurrence, greater physical symptoms, and poor sexual satisfaction.42 At 1 year, all patients, both those using complementary and alternative therapies and those using traditional methods of care, experienced an improvement in quality of life.

For patients who do not meet the criteria for clinical depression and have no interest in or access to support groups, it is worth remembering there are other interventions that can facilitate adjustment and diminish symptoms of anxiety. Expressive writing, music, or art therapy and other activity-based therapies may provide the necessary vehicles for self-expression.

Conclusion

Depression clearly affects patients with cancer, and establishing the depression diagnosis is the first step toward progress in treatment. Despite the challenges, diagnosis is possible by establishing that the symptoms of depression are negatively impacting patients' abilities to cope with their circumstances and maintain balance in their lives. It is critical not only to make the diagnosis of depression but also to strongly encourage patients to seek treatment, either through pharmacologic or nonpharmacologic means. While we make every effort to eradicate our patients' malignancies, we owe it to them to work just as diligently to improve their daily lives by treating associated depression.

Acknowledgments

We thank Dr. Donna Greenberg and Dr. William Pirl for their thoughtful review of this manuscript and helpful comments.

References2

1 W.F. Pirl, Evidence report on the occurrence, assessment, and treatment of depression in cancer patients, J Natl Cancer Inst Monogr 32 (2004), pp. 32–39. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)

2 J.S. McDaniel, D.L. Musselman, M.R. Porter, D.A. Reed and C.B. Nemeroff, Depression in patients with cancer: diagnosis, biology, and treatment, Arch Gen Psychiatry 52 (1995), pp. 89–99. View Record in Scopus | Cited By in Scopus (339)

3 R.W. Trijsburg, F.C.E. Van Knippenberg and S.E. Rijpma, Effects of psychological treatments on cancer patients: a critical review, Psychosom Med 54 (1992), pp. 489–517. View Record in Scopus | Cited By in Scopus (171)

4 A. Gruneir, T.F. Smith, J. Hirdes and R. Cameron, Depression in patients with advanced illness: an examination of Ontario complex continuing care using the minimum data set 2.0, Palliat Support Care 3 (2005), pp. 99–105. View Record in Scopus | Cited By in Scopus (6)

5 N.S. Kadan-Lottick, L.C. Vanderwerker, S.D. Block, B. Zhang and H.G. Prigerson, Psychiatric disorders and mental health service use in patients with advanced cancer: a report from the Coping with Cancer Study, Cancer 104 (2005), pp. 2872–2881. View Record in Scopus | Cited By in Scopus (57)

6 J.L. Steel, D.A. Geller, T.C. Gamblin, M.C. Olek and B.I. Carr, Depression, immunity, and survival in patients with hepatobiliary carcinoma, J Clin Oncol 25 (2007), pp. 4526–4527.

7 W. Pirl, Depression, anxiety, and fatigue. In: B. Chabner, J. Lynch and D. Longo, Editors, Harrison's Manual of Oncology, McGraw-Hill, New York (2008), pp. 190–196.

8 A. Weissman and J. Worden, The existential plight in cancer: significance of the first 100 days, Psychiatr Med 7 (1976), pp. 1–15.

9 C. Lo, C. Zimmermann and A. Rydall et al., Longitudinal study of depressive symptoms in patients with metastatic gastrointestinal and lung cancer, J Clin Oncol 28 (18) (2010), pp. 3084–3089. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)

10 National Comprehensive Cancer Network, NCCN practice guidelines for the management of psychosocial distress, Oncology (Williston Park) 13 (1999), pp. 113–147.

11 National Comprehensive Cancer Network, NCCN clinical practice guidelines in oncology, Distress management (2010) V.1. www.nccn.org.

12 M. Fisch, Treatment of depression in cancer, J Natl Cancer Inst Monogr 32 (2004), pp. 105–111. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (45)

13 H.T. Mermelstein and L. Lesko, Depression in patients with cancer, Psychooncology 1 (1992), pp. 199–215. Full Text via CrossRef

14 American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders (4th ed.), American Psychiatric Association, Washington DC (1994).

15 M.L. Chen, C.T. Yu and C.H. Yang, Sleep disturbances and quality of life in lung cancer patients undergoing chemotherapy, Lung Cancer 62 (2008), pp. 391–400. Article |

PDF (162 K)
| View Record in Scopus | Cited By in Scopus (8)

16 J. Savard, S. Simard and J. Blanchet et al., Prevalence, clinical characteristics, and risk factors for insomnia in the context of breast cancer, Sleep 24 (2001), pp. 583–590. View Record in Scopus | Cited By in Scopus (81)

17 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)

18 O.G. Palesh, J.A. Roscoe, K.M. Mustian, T. Roth, J. Savard, S. Ancoli-Israel, C. Heckler, J.Q. Purnell, M.C. Janelsins and G.R. Morrow, Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 28 (2010), pp. 292–298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

19 I. Fras, E.M. Litin and J.S. Pearson, Comparison of psychiatric symptoms in carcinoma of the pancreas with those in some other intraabdominal neoplasms, Am J Psychiatry 123 (1967), pp. 1553–1562. View Record in Scopus | Cited By in Scopus (38)

20 R.T. Joffe, D.R. Rubinow, K.D. Denicoff, M. Maher and W.F. Sindelar, Depression and carcinoma of the pancreas, Gen Hosp Psychiatry 8 (1986), pp. 241–245. Article |

PDF (583 K)
| View Record in Scopus | Cited By in Scopus (37)

21 R.P. Morton, A.D.M. Davies, J. Baker, G.A. Baker and P.M. Stell, Quality of life in treated head and neck cancer patients: a preliminary report, Clin Otolaryngol 9 (1984), pp. 181–185. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (59)

22 , Micromedex 2.0. DrugPoint summary, interferon alfa-2b http://www.micromedex.com/2 Accessed July 1, 2010.

23 D. Spiegel, A 43-year-old woman coping with cancer, JAMA 281 (4) (1999), pp. 371–377.

24 D.F. Cella, S.M. Mahon and M.I. Donovan, Cancer recurrence as a traumatic event, Behav Med 16 (1990), pp. 15–22. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (126)

25 H.M. Chochinov, K.G. Wilson, M. Enns and S. Lander, “Are you depressed?”: Screening for depression in the terminally ill, Am J Psychiatry 154 (1997), pp. 674–676. View Record in Scopus | Cited By in Scopus (225)

26 E. Bruera, L. Driver and E. Barnes et al., Patient controlled methylphenidate for cancer-related fatigue: a preliminary report, Proc Annu Meet Am Soc Clin Oncol 22 (2003), p. 737.

27 J. Homsi, K.A. Nelson and N. Sarhill et al., A phase II study of methylphenidate for depression in advanced cancer, Am J Hosp Palliat Care 18 (2001), pp. 403–407. View Record in Scopus | Cited By in Scopus (49)

28 G.R. Kalash, Psychotropic drug metabolism in the cancer patient: clinical aspects of management of potential drug interactions, Psychooncology 7 (1998), pp. 307–320. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)

29 E.C. Devine and S.K. Westlake, The effects of psychoeducational care provided by adults with cancer: meta-analysis of 116 studies, Oncol Nurs Forum 22 (1995), pp. 1369–1381. View Record in Scopus | Cited By in Scopus (192)

30 T.J. Meyer and M.M. Mark, Effects of psychosocial interventions with adult cancer patients: a meta-analysis of randomized experiments, Health Psychol 14 (1995), pp. 101–108. Abstract |

PDF (3776 K)
| Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (475)

31 S.A. Newell, R.W. Sanson-Fisher and N.J. Savolainen, Systematic review of psychological therapies for cancer patients: overview and recommendations for the future, J Natl Cancer Inst 94 (2002), pp. 558–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (201)

32 T. Sheard and P. Maguire, The effect of psychological interventions on anxiety and depression in cancer patients; results of two meta-analyses, Br J Cancer 80 (1999), pp. 1770–1780. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (210)

33 S. Williams and J. Dale, The effectiveness of treatment for depression/depressive symptoms in adults with cancer: a systematic review, Br J Cancer 94 (2006), pp. 372–390. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (60)

34 D. Spiegel, J.R. Bloom, H.C. Kraemer and E. Gottheil, Effect of psychosocial treatment on survival of patients with metastatic breast cancer, Lancet 2 (1989), pp. 888–891. Article |

PDF (618 K)
| View Record in Scopus | Cited By in Scopus (1113)

35 L.F. Berkman and S.L. Syme, Social networks, host resistence, and mortality: a nine year follow-up study of Alameda County residents, Am J Epidemiol 109 (1979), pp. 186–204. View Record in Scopus | Cited By in Scopus (1297)

36 D.P. Funch and J. Marshall, The role of stress, social support and age in survival from breast cancer, J Psychosom Res 27 (1983), pp. 77–83. Abstract |

PDF (553 K)
| View Record in Scopus | Cited By in Scopus (62)

37 D.C. Ganster and B. Victor, The impact of social support on mental and physical health, Br J Med Psychol 61 (1988), pp. 17–36. View Record in Scopus | Cited By in Scopus (17)

38 F.I. Fawzy, N. Cousins, N.W. Fawzy, M.E. Kemeny, R. Elashoff and D. Morton, A structured psychiatric intervention for cancer patients: I: Changes over time in methods of coping and affective disturbance, Arch Gen Psychiatry 47 (1990), pp. 720–725. View Record in Scopus | Cited By in Scopus (331)

39 D. Spiegel and J.R. Bloom, Group therapy and hypnosis reduce metastatic breast carcinoma pain, Psychosom Med 45 (1983), pp. 333–339. View Record in Scopus | Cited By in Scopus (192)

40 T. Gansler, C. Kaw, C. Crammer and T. Smith, A population-based study of prevalence of complementary methods use by cancer survivors: a report from the American Cancer Society's studies of cancer survivors, Cancer 113 (2008), pp. 1048–1057. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)

41 M.A. Richardson, T. Sanders, J.L. Palmer, A. Greisinger and S.E. Singletary, Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology, J Clin Oncol 18 (13) (2000), pp. 2505–2514. View Record in Scopus | Cited By in Scopus (407)

42 H.J. Burstein, S. Gelber, E. Guadagnoli and J.C. Weeks, Use of alternative medicine by women with early-stage breast cancer, N Engl J Med 340 (22) (1999), pp. 1733–1739. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (352)

Conflicts of interest: None to disclose.

Correspondence to: Alicia K. Morgans, MD, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114; telephone: (617) 724-4000; fax: (617) 643-0798


2 PubMed ID in brackets

Publications
Publications
Topics
Article Type
Display Headline
Recognizing Depression in Patients with Cancer
Display Headline
Recognizing Depression in Patients with Cancer
Sections
Article Source

PURLs Copyright

Inside the Article

Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse

Article Type
Changed
Fri, 01/04/2019 - 11:02
Display Headline
Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse

How we do it

Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse

Elizabeth Manchen RN, MS, OCN

,
, Caroline Robert MD, PhD and Camillo Porta MD

Received 12 August 2010; 

accepted 22 November 2010. 

Available online 13 February 2011.

Abstract

One significant toxicity associated with the anticancer tyrosine kinase inhibitors (TKIs) is hand–foot skin reaction (HFSR). We provide an overview of HFSR, emphasizing experience-based prevention techniques and nursing management strategies from the viewpoints of a medical oncologist, a dermatologist, and an oncology nurse. Supporting data include (1) published preclinical and phase I–III clinical studies and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib. HFSR has been reported in up to 60% of patients treated with sorafenib or sunitinib. TKI-induced HFSR may lead to dose reductions or treatment interruptions and reduced quality of life. Symptoms of TKI-associated HFSR can be managed by implementing supportive measures and aggressive dose modification. Patients educated about HFSR can work with their health-care teams to proactively detect and help manage this cutaneous toxicity, thus preventing or reducing the severity of TKI-associated HFSR. Successful prevention and management of TKI-associated HFSR can help to ensure that patients achieve optimal therapeutic outcomes. Implementation of such measures may increase the likelihood that therapy is continued for the appropriate interval at an appropriate dose for each patient. Optimal management of TKI-associated HFSR is predicated on establishing appropriate partnerships among medical oncologists, dermatologists, oncology nurses, and patients.

Article Outline

The Medical Oncologist's Viewpoint
Molecularly Targeted Agents
Characteristics of Hand–Foot Skin Reaction
Grading HFSR

The Dermatologist's Viewpoint
Incidence and Severity of HFSR With TKI
Management Strategies

The Oncology Nurse's Viewpoint

Summary

Acknowledgements

References

We are living in an era of “molecularly targeted therapy.” This targeted approach has developed as advances in science have led to a more detailed understanding of the inner workings of the cell, both in health and in illness. Once a molecular pathway has been implicated in the development and progression of cancer, modulators can be developed to intervene in this pathway. The goal is to target the cells and pathways specifically involved in the disease process, thereby leaving the normal processes undisturbed. The era of targeted therapy began in the late 1990s when the first targeted anticancer agents received regulatory approval (Figure 1). The targeted therapy armamentarium has since grown, creating a therapeutic landscape in which the chemical destruction of tumors with chemotoxic agents has been either expanded upon or replaced with agents designed to target carcinogenic processes.



Figure 1. 

Progress in the Medical Treatment of Cancer

This article reviews the mechanism of action, clinical trial results, and adverse effects of two molecularly targeted anticancer agents, the tyrosine kinase inhibitors (TKIs) sorafenib (Nexavar®; Bayer HealthCare Pharmaceuticals, Montville, NJ, and Onyx Pharmaceuticals, Emeryville, CA) and sunitinib (Sutent®; Pfizer Pharmaceuticals, New York, NY). This article specifically focuses on the diagnosis and management of TKI-associated hand–foot skin reaction (HFSR) from the perspectives of the medical oncologist, the dermatologist, and the oncology nurse. Data were derived from (1) published reports of preclinical and phase I–III clinical studies of sorafenib and sunitinib and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib.

The Medical Oncologist's Viewpoint

Molecularly Targeted Agents

Molecularly targeted therapies are directed at specific mechanisms involved in cell division, invasion, and metastasis, as well as in cell survival mediated by avoidance of apoptosis and resistance to conventional treatments. Clinical trials in several cancer types have shown that these TKIs can inhibit these activities of cancer cells by either cytostatic or cytotoxic mechanisms.1 However, the ability of these agents to inhibit multiple cancer cell pathways via novel mechanisms of action may explain, at least in part, their apparent direct toxic effects.2 These include adverse events that, from a medical viewpoint, must be anticipated, promptly recognized, and properly treated. Doing so can help minimize disruption to the patient's quality of life and may reduce the need for dose reduction or treatment interruption.1

Both sorafenib and sunitinib are orally administered, small-molecule inhibitors of multiple kinases, some of which are common to both agents (Figure 2).3 Sorafenib has known effects on tumor-cell proliferation and angiogenesis. Its antiproliferative effects are exerted via inhibition of serine/threonine kinases of the RAF/MEK/ERK signaling pathway (also called the MAP-kinase pathway) that is found within tumor cells; specifically, sorafenib targets wild-type RAF gene products (CRAF, BRAF) and mutant BRAF. The antiangiogenic effects of sorafenib are exerted via its inhibition of extracellular vascular endothelial growth factor (VEGF) receptors 2 and 3 (VEGFR-2 and VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-β), which is found mainly in the tumor vasculature. Sorafenib also exerts broad-spectrum activity against the stem-cell growth factor receptor (c-KIT), FMS-like tyrosine kinase 3 (Flt3), and the receptor encoded by the ret proto-oncogene (RET).[4], [5], [6] and [7] Sunitinib has demonstrated effects on the growth, pathologic angiogenesis, and metastatic progression of cancer by inhibiting PDGFR-α and -β; VEGFR-1, -2, and -3; and colony-stimulating factor receptor (CSF-1R), c-KIT, Flt3, and RET.8

 

 



Figure 2. 

Mechanisms of Action of Sorafenib and Sunitinib

Sorafenib and sunitinib specifically recognize and inhibit c-KIT, VEGFR, PDGFR-β, and Flt3 receptor tyrosine kinases. Sorafenib also inhibits RAF, a serine/threonine kinase involved in the RAF/MEK/ERK kinase pathway

c-KIT = stem-cell growth factor receptor (a cytokine receptor expressed on the surface of hematopoietic stem cells as well as other cell types); ERK = extracellular signal–regulated kinase; Flt3 = FMS-like tyrosine kinase 3 (a cytokine receptor expressed on the surface of hematopoietic progenitor cells); Flt3L = FMS-like tyrosine kinase 3 ligand (Flt3 ligand); MEK = (MAPK/ERK) kinase; PDGF-ΒΒ = platelet-derived growth factor BB; PDGFR-β = platelet-derived growth factor receptor beta; RAF = a gene that encodes for a protein kinase (Raf1) that functions in the mitogen-activated protein kinase/extracellular signal–regulated kinase (MAPK/ERK) signal-transduction pathway as part of a protein kinase cascade; RAS = a superfamily of genes that encode small GTPases involved in cellular signal transduction; SCF = stem-cell factor; VEGF = vascular endothelial growth factor; VEGFR = vascular endothelial growth factor receptor

Adapted with permission from Lacouture et al3

Sorafenib was approved for the treatment of advanced renal cell carcinoma (RCC) in 2005 and for unresectable hepatocellular carcinoma (HCC) in 2007. The efficacy of sorafenib in 903 patients with advanced RCC was demonstrated in the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial (TARGET), the largest phase III trial ever conducted in the second-line setting in patients with advanced RCC. Sorafenib significantly enhanced median progression-free survival (PFS) compared with placebo (24 vs 12 weeks),9 which led to early termination of the study and crossover from placebo to active drug. A preplanned analysis, which did not include patients who received placebo (who had crossed over to active treatment), ultimately demonstrated that sorafenib significantly prolonged overall survival (OS).10 Furthermore, 84% of sorafenib-treated patients experienced a clinical benefit, defined as objective response or disease stabilization.9 These results have been confirmed in larger, “real-world” patient populations in expanded-access programs conducted in North America (n = 2504)11 and the European Union (n = 118).12

Definitive data supporting the efficacy of sorafenib in HCC were provided by the randomized, double-blind, placebo-controlled Sorafenib CCC Assessment Randomized Protocol (SHARP) trial, the largest phase III trial ever conducted in patients with advanced HCC (n = 599) and the first phase III study to demonstrate a significant survival advantage with a systemic treatment in advanced HCC. In this trial, patients treated with sorafenib experienced a 44% increase in median OS (10.7 vs 7.9 months, hazard ratio [HR] = 0.69, 95% confidence interval [CI] 0.55–0.87, P < 0.001) and a 73% prolongation in median time to radiographic progression (5.5 vs 2.8 months, P < 0.001) compared with patients who received placebo.13 These results were confirmed in a separate phase III, randomized, double-blind, placebo-controlled study conducted in 226 patients from the Asia-Pacific region with advanced HCC.14 In this trial also, sorafenib significantly prolonged median OS (6.5 vs 4.2 months, HR = 0.68, 95% CI 0.50–0.93, P = 0.014) and time to progression (TTP) (2.8 vs 1.4 months, HR = 0.57, 95% CI 0.42–0.79, P = 0.0005) compared with placebo.

Sunitinib received approval in 2006 for use in patients with gastrointestinal stromal tumor (GIST) whose disease is refractory to imatinib (Gleevec®; Novartis Pharmaceuticals, East Hanover, NJ) or who are intolerant to the drug and in those with advanced RCC. Approval of sunitinib for the treatment of GIST was based on data from a randomized, placebo-controlled, phase III trial of 312 patients with imatinib-refractory GIST.15 In that study, sunitinib treatment increased median PFS (24.1 vs 6.0 weeks, HR = 0.33, 95% CI 0.24–0.47, P < 0.0001) and median TTP (27.3 vs 6.4 weeks, HR = 0.33, 95% CI 0.23–0.47, P < 0.0001) compared with placebo. The trial was unblinded early when a planned interim analysis revealed significantly longer TTP with sunitinib than with placebo. A subsequent analysis showed that median OS with sunitinib was about twice that with placebo (73.9 vs 35.7 weeks, P < 0.001).16 In an ongoing, worldwide treatment-use program to provide expanded access to sunitinib for patients with advanced GIST intolerant of or resistant to imatinib,17 sunitinib treatment resulted in a median estimated TTP of 41 weeks and a median estimated OS of 75 weeks in the population analyzed (n = 1,117).

A separate phase III randomized controlled trial was conducted in 750 patients with advanced RCC and no history of systemic therapy for RCC.[18] and [19] The active comparator in this trial was interferon-alfa (IFN-α). Compared with IFN-α, sunitinib significantly increased median PFS (11 vs 5 months, HR = 0.539, 95% CI 0.451–0.643, P < 0.001) and was associated with a greater objective response rate (47% vs 12%, P < 0.001). Median OS was greater in the sunitinib group (26.4 vs 21.8 months), but the difference was not significant (P = 0.051). Data from expanded-access programs in patients with RCC and GIST support the phase III trial data for sunitinib.20 Efficacy data for sorafenib and sunitinib are summarized in Table 1.[9], [10], [11], [12], [13], [14], [15], [17], [18] and [20]

 

 

Table 1. Summary of Efficacy Data for Sorafenib and Sunitinib

DRUG, DISEASE, AND STUDYEFFICACY DATA
Sorafenib
Advanced RCC
Phase III TARGET[9] and [10]

• Largest phase III trial ever conducted in second-line setting in patients with advanced RCC

• Median PFS was 5.5 months in sorafenib group vs 2.8 months in placebo group (P < 0.001)

• 28% improvement in OS vs placebo (HR = 0.72, P = 0.02)

• Significant prolongation of OS (HR = 0.78, 95% CI 0.62–0.97, P = 0.029)

• Clinical benefit (CR + PR + SD) in 84% of patients

Expanded-access programs[11] and [12]Data from expanded-access programs in community-based populations (NA- and EU-ARCCS) were consistent with data from TARGET
Unresectable HCC
Phase III SHARP trial13

• First phase III trial to demonstrate a significant survival advantage for a systemic therapy in advanced HCC

• Median OS was 10.7 months in sorafenib group vs 7.9 months in placebo group (HR = 0.69, 95% CI 0.55–0.87, P < 0.001)

• Median TTRP was 5.5 months in sorafenib group vs 2.8 months in placebo group (HR = 0.58, 95% CI 0.45–0.74, P < 0.001)

• Disease control rate (CR + PR + SD) was 43% in sorafenib group vs 32% in placebo group (P = 0.002)

Phase III Asia-Pacific trial14

• Median OS was 6.5 months in sorafenib group vs 4.2 months in placebo group (HR = 0.68, 95% CI 0.50–0.93, P = 0.014)

• Median TTP was 2.8 months in sorafenib group vs 1.4 months in placebo group (HR = 0.57, 95% CI 0.42–0.79, P = 0.0005)

Sunitinib
Advanced RCC
Phase III registration trial18

• Median PFS was 11 months in sunitinib group vs 5 months in interferon-α group (HR = 0.539, 95% CI 0.451–0.643, P < 0.001)

• Objective response rate (CR + PR) was 47% in sunitinib group vs 12% in interferon-α group (P < 0.001)

• Median OS was 26.4 months in sunitinib group vs 21.8 months in interferon-α group (P = 0.051)

Expanded-access program20

• In a broad population of patients with metastatic RCC who were treated with sunitinib:

 – Median PFS was 10.9 months

 – Median OS was 18.4 months

Imatinib-resistant GIST
Phase III registration trial15

• Median TTP was 27.3 weeks in sunitinib group vs 6.4 weeks in placebo group (HR = 0.33, 95% CI 0.23–0.47, P < 0.0001)

• Median PFS was 24.1 weeks in sunitinib group vs 6.0 weeks in placebo group (HR = 0.33, 95% CI 0.24–0.47, P < 0.0001)

• 16% of sunitinib-treated patients were progression-free for at least 26 weeks compared with 1% of those who received placebo

Expanded-access program17

• In a broad population of patients with imatinib-resistant GIST who were treated with sunitinib:

 – Estimated median TTP was 41 weeks

 – Estimated median OS was 75 weeks

CI = confidence interval; CR = complete response; EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; HR = hazard ratio; NA-ARCCS = North American ARCCS; OS = overall survival; PFS = progression-free survival; PR = partial response; RCC = renal cell carcinoma; SD = stable disease; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial; TTP = time to progression; TTRP = time to radiologic progression


Characteristics of Hand–Foot Skin Reaction

Data from the clinical trials for sorafenib and sunitinib indicate that both agents are generally well-tolerated; common treatment-related adverse reactions include diarrhea, alopecia, nausea, fatigue, rash, and hypertension, as well as palmar–plantar erythrodysesthesia (PPE) syndrome, also known as hand–foot skin reaction (HFSR) (Table 2).[10] and [19] HFSR is a dermatologic toxicity that has been reported in 14%–62% of patients treated with sorafenib or sunitinib (Table 3).[9], [11], [12], [13], [14], [15], [17], [18], [20], [21], [22], [23], [24] and [25] In general, the term HFSR refers to a group of signs and symptoms affecting the hands and feet of patients taking sorafenib, sunitinib, or, to a lesser extent, other TKIs such as pazopanib (Votrient™; GlaxoSmithKline, Research Triangle Park, NC)[26] and [27] and axitinib (AG013736).[28], [29], [30] and [31]

Table 2. Selected Common Adverse Events in Patients Treated with Sorafenib (n = 452) or Sunitinib (n = 375) in Phase III Registration Trials (Updated and Final Results)[10] and [19]

ADVERSE EVENT
SORAFENIB 400 MG BID
SUNITINIB 50 MG QD
ALL GRADES (%)GRADE 3/4 (%)ALL GRADES (%)GRADE 3/4 (%)
Diarrhea483619
Rash411242
Hand–foot skin reaction336299
Alopecia310120
Fatigue2935411
Nausea19<1525
Hypertension1743012
Dry skin13021<1
Vomiting121314
Mucositis50262

Table 3. Rates of Hand–Foot Skin Reaction in Clinical Trials of Sorafenib and Sunitinib

REFERENCESTUDYALL GRADES (%)GRADE 3 (%)GRADE 4 (%)
Sorafenib
 9Phase III TARGETa306 (grade 3/4)
 13Phase III SHARPa2180
 11NA-ARCCS, first-linea19 (≥2)11 (grade 3/4)
 11NA-ARCCS, second-linec17 (≥2)8 (grade 3/4)
 12EU-ARCCSa4712 (grade 3/4)
 14Phase III Asia-Pacifica4511 (grade 3/4)
 24Phase II randomized discontinuation trial in advanced RCCb62130
 25Phase II study in advanced HCCb3150
 21Phase II, uncontrolled study in relapsed/refractory NSCLCa3710 (grade 3/4)
Sunitinib
 18Phase III registration trial in advanced RCCa2050
 20Expanded access program in advanced RCCc5 (grade 3/4)
 15Phase III registration trial in imatinib-resistant GISTa1440
 [17] and [22]Expanded access program in imatinib-resistant GISTcN/A8 (grade 3/4)
 23Phase II trial of second-line treatment in advanced RCCa1570

EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; N/A = data not available; NA-ARCCS = North American ARCCS; NSCLC = non-small-cell lung cancer; RCC = renal cell carcinoma; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial

a Used version 3.0 of National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE)b Used version 2.0 of NCI-CTCAEc Version of NCI-CTCAE used not specified

 

 

HFSR is typically characterized by redness, marked discomfort, swelling, and tingling in the palms of the hands and/or soles of the feet.32 HFSR can be painful enough to interfere profoundly with activities of daily living (ADLs). In fact, patients may report symptoms after as few as 2 weeks on TKI therapy, at which point they may present to the health-care provider (HCP) wearing slippers, unable to walk, and having difficulty in performing ADLs such as eating, dressing, and bathing.[1] and [33] Although HFSR can lead to TKI dose modification or treatment discontinuation, preventive measures can be taken before TKIs are initiated to reduce the likelihood of HFSR. In addition, early treatment of symptoms may prevent HFSR from progressing to the point at which the patient's ability to receive the full potential benefit of therapy is compromised.[3], [34], [35] and [36]

Signs and symptoms of HFSR may appear concomitantly or sequentially and can affect both hands and both feet. Although symptoms are most prominent on the palms and soles, other areas of the hands and feet may also be involved, including the tips of the fingers and toes, the heels, and metatarsophalangeal skin; areas of flexure; and skin overlying the metacarpophalangeal and interphalangeal joints.3 These “pressure areas” are where the most severe symptoms are typically seen. Common symptoms include dysesthesia and paresthesia, described as “tingling, prickling, or creeping sensations” and/or sensitivity or intolerance to hot or warm objects (which may occur before other symptoms are apparent); erythema; edema; hyperkeratosis; and dry and/or cracked skin.[1] and [34] Actual HFSR lesions are described as tender and scaling, with a peripheral halo of erythema, yellowish and hyperkeratotic plaques, or callous-like blisters (which usually do not contain fluid), typically localized to areas of pressure.[3] and [35] Desquamation, particularly with sunitinib treatment, may also be present.37

Since both sorafenib and sunitinib inhibit the VEGFRs, PDGFRs, c-KIT, and Flt3,38 it is likely that inhibition of one or more of these receptors and/or pathways plays a role in HFSR development.36 Differences in the relative appearance of HFSR symptoms are dependent on whether sorafenib or sunitinib is used. Sunitinib use is more often associated with desquamation, whereas sorafenib is more often associated with areas of hyperkeratosis, particularly formation of thick calluses on the soles of the feet.37 The timing of the first appearance of symptoms may also vary according to the TKI used. HFSR usually develops within the first 2–4 weeks of treatment with a TKI and almost always within the first 6 weeks.35 However, because the severity of HFSR appears to be dose-dependent,3 signs and symptoms may present later rather than sooner in patients treated with sunitinib. This is likely due to the recommended sunitinib dosing schedule, which incorporates a 2-week period during which no drug is administered. Although HFSR frequently decreases in intensity during treatment, even without dose modifications or treatment interruption, prompt treatment of HFSR is recommended to prevent rapid progression. Early symptoms can usually be resolved easily by appropriate treatment, which often allows continuation of full-dose therapy for the prescribed length of time.

It is important to note what HFSR is not. TKI-associated HFSR is not the same clinical entity as the hand–foot syndrome (HFS) traditionally seen with cytotoxic agents such as infusional 5-fluorouracil (5-FU); capecitabine, the oral prodrug of 5-FU; and pegylated liposomal doxorubicin. Although HFSR and HFS share several clinical and pathological aspects—each previously has been called “acral erythema” and “PPE”—they clearly are not the same clinical or pathologic entity. HFSR is neither an allergic reaction to a drug nor an indication that a patient may be intolerant to a drug. Finally, HFSR does not indicate drug efficacy, as may be the case with skin rash in patients with non-small-cell lung cancer treated with erlotinib.[3] and [39]

Grading HFSR

In published reports, the severity of HFSR is usually graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE),32 a three-grade classification system. However, modified criteria are considered by some to be a better fit for routine clinical practice.1 What distinguishes the modified criteria from the NCI criteria (version 4.02) is the inclusion of HFSR-specific clinical characteristics plus certain patient-defined considerations used to categorize severity. The modified criteria expressly define the degree to which HFSR discomfort affects the patient's normal activities, an improvement over version 4.02 used alone. The NCI-CTCAE version 4.02 criteria, the modified criteria, and corresponding patient photographs are presented in Figure 3.[1] and [32]

 

 



Figure 3. 

National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE, version 4.02)32 and Modified Grading Criteria for Hand–Foot Skin Reaction1

Note that grade-4 toxicity is not contemplated in either system

Photographs reproduced with permission

Recommendations for the treatment of grade 1 HFSR include early and appropriate dermatologic management and active collaboration among HCPs.40

The Dermatologist's Viewpoint

Although the exact pathogenesis of HFSR has not been fully elucidated, research into its cause(s) is ongoing. Theoretically, traditional HFS is thought to be due to the direct toxic effects of drugs or their ability to invoke a “host-vs-host” response. In contrast, a unique mechanism has been proposed for TKI-associated HFSR: simultaneous blockage of VEGFRs and PDGFRs.2

Three histopathologic features have been found to predominate in HFSR: dyskeratotic keratinocytes at various states of necrosis (Figure 441), basal layer vacuolar degeneration, and mild perivascular or lichenoid lymphocyte-predominant infiltrate.2 Immunohistochemistry with a variety of skin-cell markers has shown a significant modification of normal maturation of keratinocytes, which are often apoptotic. Minor modifications of blood vessels are also seen, but no signs of intense vasculitis are evident. This is important because HFSR is suspected of being a “class effect” of TKIs that target VEGFRs. HFSR is not seen in patients treated with single-agent bevacizumab, and the lack of histologic evidence of significant damage to blood vessels suggests that HFSR does not result from the general inhibition of angiogenesis. A retrospective analysis found that HFSR rates were higher when patients were treated with sorafenib and bevacizumab in combination, supporting the hypothesis that HSFR is due to the anti-VEGF properties of sorafenib.42 Other possible causes of HFSR include activation by a ligand other than VEGF and/or inhibition of one of the other protein targets inhibited by both sorafenib and sunitinib.[3] and [35]



Figure 4. 

Layers of the Epidermis

The epidermis is composed of a very sophisticated arrangement of keratinocytes, which originate as stem cells in the stratum germinatum (not shown). The stem cells constantly multiply, creating daughter cells that progressively mature over approximately 28 days and move to the surface of the epidermis. As they move, they change their function and shape. In the stratum spinosum, interactions between cells resemble spines; mature cells move through the stratum granulosum, which has a very important secretory function, until finally the cells reach the stratum corneum and die. Thus, the stratum corneum is the layer of skin containing dead skin cells that have lost their nuclei; it is the part of the epidermis that ensures the barrier function of skin and is the layer most affected by HFSR. The layer beneath the epidermis is called the “papillary dermis.” The papillary dermis contains nerves and blood vessels and supplies the epidermis with nutrients. The fibroblasts and fibers located here give skin its strength and resistance

Adapted with permission from Gawkrodger41


Incidence and Severity of HFSR With TKI

To determine the incidence and severity of HFSR specific to sorafenib, a double-blind, prospective, dermatologic substudy was performed in patients enrolled in the phase III TARGET trial.35 Eighty-five patients with RCC were randomized to receive either sorafenib (n = 43) or placebo (n = 42). Dermatologic examinations were performed before and during treatment. Ninety-one percent of sorafenib-treated patients experienced at least one cutaneous reaction compared with 7% of those in the placebo group. A variant of HFSR clinically distinct from chemotherapy-induced HFS was observed in 60% of sorafenib-treated patients. Reversible grade 3 HFSR leading to dose reduction occurred in two sorafenib-treated patients. Additional cutaneous reactions were facial erythema, scalp dysesthesia, alopecia, and subungual splinter hemorrhages.

HFSR (of any grade) has been shown to occur in approximately 30% of patients treated with sorafenib and 20% of those who received sunitinib in clinical studies.43 Grade 3/4 HFSR has been observed in approximately 6% of sorafenib-treated and 5% of sunitinib-treated patients. HFSR was not reported in a phase II study of 142 patients with relapsed or refractory soft-tissue sarcoma treated with pazopanib.44 In a phase III randomized, double-blind, placebo-controlled trial of pazopanib in patients with advanced RCC, the incidence of HFSR was <10%, while the incidence of grade 3/4 HFSR was <1%. Potential differences may be explained by variations in the potency and selectivity of the TKIs.27

Management Strategies

Our work at the Dermatology Center at the Gustave-Roussy Institute has shown that early intervention against the dermatologic adverse effects of these TKIs can inhibit patient progression to a more serious form of HFSR.[34] and [38]

Effective management of HFSR can begin prior to initiation of treatment with sorafenib or sunitinib. Patients should be advised to remove any preexisting hyperkeratotic areas or calluses, keep skin well-moisturized with appropriate creams, and cushion pressure points with cotton socks, soft shoes, and/or insoles. Dose modification is typically not required for grade 1 HFSR; symptomatic treatments should be employed instead.

If HFSR symptoms progress to grade 2 or 3, with pain and a decrease in quality of life, the dose of sorafenib or sunitinib can be modified until symptoms recede, after which the patient can be brought back to the full dose. Very often, the patient can tolerate the full-dose treatment simply by decreasing the dose briefly.3 A recommended dose-modification scheme is shown in Figure 5.3

 

 



Figure 5. 

Management of Hand–Foot Skin Reaction by Grade

MKI = multikinase inhibitor

Adapted with permission from Lacouture et al3

TKIs are being studied in patients with additional types of tumor, possibly in the adjuvant setting, as well as in combinations. Because these drugs are administered orally, with a decreased (compared with conventional cytotoxic agents) frequency of nurse– or doctor–patient interactions, patients must be very well-informed of any potential toxicities with the TKIs.

At present, there are no evidence-based treatment guidelines for the prevention or management of HFSR. However, HCPs most involved in the day-to-day care of patients with HFSR have made great progress in establishing preventive and treatment strategies and in identifying ancillary products likely to decrease the incidence and/or severity of symptoms. Prevention, which includes preventing HFSR entirely as well as preventing progression from its initial appearance, is a key component of HFSR management.

The Oncology Nurse's Viewpoint

The nurse's viewpoint begins with patient education and empowerment. The goal is to prevent adverse effects from occurring while managing any adverse effects that do occur so that the patient has the best chance of staying on anticancer therapy. This requires a strong partnership between the HCP team and the patient. Although not all cases of HFSR can be prevented, experience suggests that symptom incidence and severity can be alleviated by educating patients to recognize the signs and symptoms of HFSR and report these to their HCPs. HFSR typically occurs early in the course of therapy, so it is prudent to be especially vigilant during the first 6 weeks. Providing the patient with a brochure about HFSR to refer to at home may facilitate identification of HFSR.

To address the lack of evidence-based guidelines to prevent or treat HFSR, an international, interdisciplinary expert panel has provided a set of consensus recommendations for the management of TKI-associated HFSR.45 One component of these recommendations can be phrased simply for the patients as a “3C” approach to management: control calluses, comfort with cushions, and cover with creams.

Prior to treatment, the patient should receive a full-body examination, with emphasis on the condition of the hands and feet. Evaluation should be performed by a qualified HCP who can determine whether there are physical conditions that may predispose a patient to areas of increased friction or rubbing. For all patients, especially those with comorbid conditions (eg, diabetes, poor circulation), a pretreatment pedicure is highly recommended. Patients should also be educated on the proper use of tools (eg, a pumice stone) to aid in callus removal. Such tools are considered beneficial because patients can control the frequency of their use and the extent of skin removed. However, because areas of hyperkeratosis are often extremely tender and painful, patients are cautioned against overuse of these tools, including the aggressive “paring” or “cutting” of callused areas. Finally, patients should be advised of the need for clean tools to guard against infection.

Other protective measures include the use of thick cotton gloves and/or socks, which may also help the skin to retain moisture, and avoidance of warm and/or hot water or objects, tight-fitting shoes, or other items that may rub, pinch, or cause friction in affected areas. Tender areas, pressure points, and pressure-sensitive areas of the hands and feet should be protected. For example, weight lifters might be advised to wear gloves. These recommendations hold true both before and after development of HFSR.[3], [33] and [35] Well-padded but nonconstrictive footwear should be worn, and the use of insole cushions or inserts (eg, silicone or gel) should be encouraged. Foot soaks with lukewarm water and magnesium sulfate may be soothing. Tender areas should be protected at all times, and patients should be encouraged not to walk barefoot.

Use of over-the-counter and prescription-strength creams and moisturizers during treatment with TKIs has also been recommended (Table 4).[40], [45], [46], [47], [48] and [49] Moisturizing agents should be applied liberally, immediately after bathing. Cotton gloves and/or socks can also be worn, to help retain moisture and to provide an additional layer of protection. When applied liberally, these products soften areas of thick and hardened skin, help keep the skin pliable, and may prevent cracks or breaks in skin integrity, which could cause additional discomfort. Prescription-strength topical agents have also shown anecdotal benefit (Table 4). These topical agents are typically applied twice daily to affected areas only because they may irritate unaffected skin. Data on the use of topical/systemic corticosteroids in the treatment of HFS remain inconclusive; the literature primarily includes case studies in patients with PPE treated with chemotherapeutic agents including pegylated liposomal doxorubicin.2 Finally, a qualified HCP must always be consulted to ensure proper diagnosis and treatment of HFSR.

 

 

Table 4. Selected Skin Care Products for Use in HFSR

Adapted with permission from Anderson et al40

PRODUCTSPRODUCT INFORMATION
Over-the-counter[40], [45] and [46]
 Cetaphil® (Galderma Laboratories, Ft. Worth, TX) skin cleaners, Aveeno® (Johnson & Johnson, New Brunswick, NJ) shower gelNondeodorant, fragrance-free products
 Udderly Smooth® (Redex Industries, Salem, OH), Gold Bond® (Chattem, Chattanooga, TN), Aveeno®Thicker products with more intense moisturizing properties than basic lotions; anti-itch formulations are available
 Norwegian Formula: Smoothing Relief Anti-Itch Moisturizer (Neutrogena, Los Angeles, CA)Contains dimethicone 1%, camphor 0.1%, and lidocaine
 Norwegian Formula: Foot Cream (Neutrogena)Contains cetearyl alcohol, dimethicone, menthol, and urea
 Bag Balm® (Dairy Association, Lyndonville, VT)May provide “cooling” effect from eucalyptus
 Eucerin® (Beiersdorf, Hamburg, Germany) CreamBest used at night due to greasy formulation
 Eucerin® Dry Skin TherapyContains urea and alpha-hydroxy acid
 Aquaphor® (Beiersdorf) Healing OintmentPetrolatum 41%
 Kerasal® (Alterna, Whippany, NJ)Salicylic acid 5% exfoliates and softens skin; urea 10% moisturizes skin
 Blue Lizard® (Crown Laboratories, Johnson City, TN)UV A and B sunblock, water-resistant
Prescription47–49
 Urea 40% cream (Carmol 40®; Doak Dermatologics, Fairfield, NJ)Softens hyperkeratosis, decreases epidermal thickness, proliferation
 Tazarotene 0.1% cream (Tazorac®; Allergan Dermatology, Irvine, CA)Retinoid decreases proliferation, reduces dermal inflammation
 Fluorouracil 5% cream (Carac®; Dermik Laboratories, Bridgewater, NJ)Antifolate inhibits proliferation; has shown anecdotal benefit in certain conditions characterized by hyperkeratotic lesions on the palms and soles


Summary

The addition of molecularly targeted agents to anticancer treatment has been found to cause both common and novel adverse reactions. HFSR is being increasingly recognized as a potential dose-limiting toxicity associated with sorafenib or sunitinib treatment that can result in discomfort, pain, decreased quality of life, and premature termination of a potentially effective cancer treatment. It is important to educate patients about potential dermatologic adverse effects associated with TKIs because limiting toxicity can help avoid treatment interruptions or dose reductions while improving ADLs.

The precise pathogenic mechanism of HFSR is currently not known, and there is no evidence-based protocol for treatment of HFSR. However, the increased clinical experience with these agents has resulted in a wealth of published articles describing empiric and symptomatic approaches that appear to help to prevent and manage HFSR. Frequent communication is necessary between the physician and patient, particularly 2–4 weeks from the initiation of therapy. Symptoms of HFSR should be recognized as early as possible. Providing the patient with a brochure about HFSR to refer to at home may facilitate the early identification of HFSR.

Patients should be advised of the “3C” approach to the management of TKI-associated HFSR: control calluses, comfort with cushions, and cover with creams. Creams should be applied after bathing and before going to bed; cotton gloves and socks should be worn to keep the cream on the hands and feet during the night.

Symptoms of HFSR typically are manageable with the implementation of supportive measures. If symptoms worsen, dose modification or interruption will result in a return to grade 0/1. Many patients can successfully be rechallenged with the full dose. Observations across multiple viewpoints have consistently shown that HFSR severity can be reduced in patients who are educated about HFSR and proactive about its detection and management.

Acknowledgments

All authors contributed equally to the development of this report. Editorial support was provided by Katherine Wright, PharmD, RPh, ISD, Wrighter Medical Education and Training, West Hills, CA; John A. Ibelli, CMPP, BelMed Professional Resources, New Rochelle, NY; and John D. Zoidis, MD, Bayer HealthCare Pharmaceuticals, Montville, NJ.

References1

1 C. Porta, C. Paglino, I. Imarisio and L. Bonomi, Uncovering Pandora's vase: the growing problem of new toxicities from novel anticancer agents: The case of sorafenib and sunitinib, Clin Exp Med 7 (2007), pp. 127–134. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

2 A.D. Lipworth, C. Robert and A.X. Zhu, Hand–foot syndrome (hand–foot skin reaction, palmar–plantar erythrodysesthesia): focus on sorafenib and sunitinib, Oncology 77 (2009), pp. 257–271 [19923864]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

3 M.E. Lacouture, S. Wu and C. Robert et al., Evolving strategies for the management of hand–foot skin reaction associated with the multitargeted kinase inhibitors sorafenib and sunitinib, Oncologist 13 (2008), pp. 1001–1011 [18779536]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (57)

4 S.M. Wilhelm, L. Adnane, P. Newell, A. Villanueva, J.M. Llovet and M. Lynch, Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling, Mol Cancer Ther 7 (2008), pp. 3129–3140 [18852116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)

5 D. Auclair, D. Miller and V. Yatsula et al., Antitumor activity of sorafenib in FLT3-driven leukemic cells, Leukemia 21 (2007), pp. 439–445 [17205056]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)

6 F. Carlomagno, S. Anaganti and T. Guida et al., BAY 43-9006 inhibition of oncogenic RET mutants, J Natl Cancer Inst 98 (2006), pp. 326–334 [16507829]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)

7 S.M. Wilhelm, C. Carter and L. Tang et al., BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis, Cancer Res 64 (2004), pp. 7099–7109 [15466206]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1142)

8 J.G. Christensen, A preclinical review of sunitinib, a multitargeted receptor tyrosine kinase inhibitor with anti-angiogenic and antitumour activities, Ann Oncol 10 (2007), pp. x3–x10 [17761721]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

9 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib in advanced clear-cell renal-cell carcinoma, N Engl J Med 356 (2007), pp. 125–134 [17215530]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1425)

10 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial, J Clin Oncol 27 (2009), pp. 3312–3318 [19451442]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (123)

11 W.M. Stadler, R.A. Figlin and D.F. McDermott et al., Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in North America, Cancer 116 (2010), pp. 1272–1280 [20082451]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)

12 J. Beck, E. Bajetta and B. Escudier et al., A large open-label, non-comparative, phase III study of the multi-targeted kinase inhibitor sorafenib in European patients with advanced renal cell carcinoma, Eur J Cancer (suppl) (2007), p. 5 Abstract 4506. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

13 J.M. Llovet, S. Ricci and V. Mazzaferro et al., Sorafenib in advanced hepatocellular carcinoma, N Engl J Med 359 (2008), pp. 378–390 [18650514]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (989)

14 A.L. Cheng, Y.K. Kang and Z. Chen et al., Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial, Lancet Oncol 10 (2009), pp. 25–34 [19095497]. Article |

PDF (195 K)
| View Record in Scopus | Cited By in Scopus (279)

15 G.D. Demetri, A.T. van Oosterom and C.R. Garrett et al., Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial, Lancet 368 (2006), pp. 1329–1338 [17046465]. Article |

PDF (200 K)
| View Record in Scopus | Cited By in Scopus (630)

16 G.D. Demetri, X. Huang and C.R. Garrett et al., Novel statistical analysis of long-term survival to account for crossover in a phase III trial of sunitinib (SU) vs placebo (PL) in advanced GIST after imatinib (IM) failure, J Clin Oncol (suppl) (2008), p. 26 Abstract 10524.

17 P. Reichardt, Y. Kang and W. Ruka et al., Detailed analysis of survival and safety with sunitinib (SU) in a worldwide treatment-use trial of patients with advanced GIST, J Clin Oncol (suppl) (2008), p. 26 Abstract 10548.

18 R.J. Motzer, T.E. Hutson and P. Tomczak et al., Sunitinib versus interferon alfa in metastatic renal-cell carcinoma, N Engl J Med 356 (2007), pp. 115–124 [17215529]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1520)

19R.J. Motzer, T.E. Hutson and P. Tomczak et al., Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 3584–3590 [19487381]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (194)

20 M.E. Gore, C. Szczylik and C. Porta et al., Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial, Lancet Oncol 10 (2009), pp. 757–763 [19615940]. Article |

PDF (142 K)
| View Record in Scopus | Cited By in Scopus (72)

21 G.R. Blumenschein Jr, U. Gatzemeier and F. Fossella et al., Phase II, multicenter, uncontrolled trial of single-agent sorafenib in patients with relapsed or refractory, advanced non-small-cell lung cancer, J Clin Oncol 27 (2009), pp. 4274–4280 [19652055]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

22 H.J. Schütte, M. Schlemmer, C. Wendtner, P. Reichardt and G.D. Demetri, Sunitinib (SU) in a worldwide treatment-use trial of pts with advanced gastrointestinal stromal tumours (GIST): analysis of survival and safety data, Onkologie (suppl 4) (2008), p. 201 Abstract V582.

23 R.J. Motzer, B.I. Rini and R.M. Bukowski et al., Sunitinib in patients with metastatic renal cell carcinoma, JAMA 295 (2006), pp. 2516–2524 [16757724]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (586)

24 M.J. Ratain, T. Eisen and W.M. Stadler et al., Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma, J Clin Oncol 24 (2006), pp. 2505–2512 [16636341]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (544)

25 G.K. Abou-Alfa, L. Schwartz and S. Ricci et al., Phase II study of sorafenib in patients with advanced hepatocellular carcinoma, J Clin Oncol 24 (2006), pp. 4293–4300 [16908937]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (418)

26 T.E. Hutson, I.D. Davis and J.P. Machiels et al., Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma, J Clin Oncol 28 (2010), pp. 475–480 [20008644]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

27 C.N. Sternberg, I.D. Davis and J. Mardiak et al., Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial, J Clin Oncol 28 (2010), pp. 1061–1068 [20100962]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (100)

28 O. Rixe, R.M. Bukowski and M.D. Michaelson et al., Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study, Lancet Oncol 8 (2007), pp. 975–984 [17959415]. Article |

PDF (143 K)
| View Record in Scopus | Cited By in Scopus (132)

29 B.I. Rini, I. Tamaskar and P. Shaheen et al., Hypothyroidism in patients with metastatic renal cell carcinoma treated with sunitinib, J Natl Cancer Inst 99 (2007), pp. 81–83 [17202116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (159)

30 B.I. Rini, G. Wilding and G. Hudes et al., Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 4462–4468 [19652060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (49)

31 H.S. Rugo, R.S. Herbst and G. Liu et al., Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: pharmacokinetic and clinical results, J Clin Oncol 23 (2005), pp. 5474–5483 [16027439]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (161)

32 National Cancer Institute, National Institutes of Health, US Department of Health and Human Services, Common Terminology Criteria for Adverse Events (CTCAE) Version 4.02. NIH publication 03-5410, National Institutes of Health, Bethesda, MD (2009).

33 L.S. Wood and B. Manchen, Sorafenib: a promising new targeted therapy for renal cell carcinoma, Clin J Oncol Nurs 11 (2007), pp. 649–656 [17962173]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)

34 C. Robert, C. Mateus, A. Spatz, J. Wechsler and B. Escudier, Dermatologic symptoms associated with the multikinase inhibitor sorafenib, J Am Acad Dermatol 60 (2009), pp. 299–305 [19028406]. Article |

PDF (296 K)
| View Record in Scopus | Cited By in Scopus (34)

35 J. Autier, B. Escudier, J. Wechsler, A. Spatz and C. Robert, Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor, Arch Dermatol 144 (2008), pp. 886–892 [18645140]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)

36 M.E. Lacouture, L.M. Reilly, P. Gerami and J. Guitart, Hand foot skin reaction in cancer patients treated with the multikinase inhibitors sorafenib and sunitinib, Ann Oncol 19 (2008), pp. 1955–1961 [18550575]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

37 L.S. Wood, Managing the side effects of sorafenib and sunitinib, Community Oncol 3 (2006), pp. 558–562. View Record in Scopus | Cited By in Scopus (19)

38 C. Robert, J.C. Soria and A. Spatz et al., Cutaneous side-effects of kinase inhibitors and blocking antibodies, Lancet Oncol 6 (2005), pp. 491–500 [15992698]. Article |

PDF (533 K)
| View Record in Scopus | Cited By in Scopus (208)

39 M.W. Saif, I. Merikas, S. Tsimboukis and K. Syrigos, Erlotinib-induced skin rash: Pathogenesis, clinical significance and management in pancreatic cancer patients, JOP 9 (2008), pp. 267–274 [18469438]. View Record in Scopus | Cited By in Scopus (11)

40 R. Anderson, A. Jatoi, C. Robert, L.S. Wood, K.N. Keating and M.E. Lacouture, Search for evidence-based approaches for the prevention and palliation of hand–foot skin reaction (HFSR) caused by the multikinase inhibitors (MKIs), Oncologist 14 (2009), pp. 291–302 [19276294]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)

41 D.J. Gawkrodger, Dermatology: An Illustrated Colour Text, 4th ed, Churchill Livingstone Elsevier, Edinburgh (2008).

42 N.S. Azad, J.B. Aragon-Ching and W.L. Dahut et al., Hand–foot skin reaction increases with cumulative sorafenib dose and with combination anti-vascular endothelial growth factor therapy, Clin Cancer Res 15 (2009), pp. 1411–1416 [19228742]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

43 N. Bhojani, C. Jeldres and J.J. Patard et al., Toxicities associated with the administration of sorafenib, sunitinib, and temsirolimus and their management in patients with metastatic renal cell carcinoma, Eur Urol 53 (2008), pp. 917–930 [18054825]. Article |

PDF (843 K)
| View Record in Scopus | Cited By in Scopus (81)

44 S. Sleijfer, I. Ray-Coquard and Z. Papai et al., Pazopanib, a multikinase angiogenesis inhibitor, in patients with relapsed or refractory advanced soft tissue sarcoma: a phase II study from the European Organisation for Research and Treatment of Cancer–Soft Tissue and Bone Sarcoma Group (EORTC study 62043), J Clin Oncol 27 (2009), pp. 3126–3132 [19451427]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)

45 L.S. Wood, H. Lemont and A. Jatoi et al., Practical considerations in the management of hand–foot skin reaction caused by multikinase inhibitors, Community Oncol 7 (2010), pp. 23–29. View Record in Scopus | Cited By in Scopus (3)

46 P. Esper, D. Gale and P. Muehlbauer, What kind of rash is it?: Deciphering the dermatologic toxicities of biologic and targeted therapies, Clin J Oncol Nurs 11 (2007), pp. 659–666 [17962174]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

47 I. Hagemann and E. Proksch, Topical treatment by urea reduces epidermal hyperproliferation and induces differentiation in psoriasis, Acta Derm Venereol 76 (1996), pp. 353–356 [8891006]. View Record in Scopus | Cited By in Scopus (41)

48 M. Duvic, A.T. Asano, C. Hager and S. Mays, The pathogenesis of psoriasis and the mechanism of action of tazarotene, J Am Acad Dermatol 39 (1998), pp. S129–S133 [9777790]. Article |

PDF (205 K)
| View Record in Scopus | Cited By in Scopus (24)

49 W. Scheithauer, J. McKendrick and S. Begbie et al., Oral capecitabine as an alternative to i.v. 5-fluorouracil-based adjuvant therapy for colon cancer: safety results of a randomized, phase III trial, Ann Oncol 14 (2003), pp. 1735–1743 [14630678]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (172)

 

 

Conflicts of interest: For E. M., none to disclose. C. R. has participated in advisory boards for Bayer, Roche, Pfizer, Astrazeneca, and GSK in the field of melanoma treatment and the management of the cutaneous side effects of anticancer agents. C. P. has acted as a paid adviser or speaker for Bayer Schering Pharma, Pfizer Oncology, Hoffman La Roche, Novartis Pharma, GSK, and Wyeth Pharmaceuticals and has received research funding from Bayer Schering Pharma and Novartis Pharma.

Correspondence to: Elizabeth Manchen, RN, MS, OCN, Section of Hematology/Oncology, University of Chicago Medical Center, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637; telephone: (773) 702–4135


1 PubMed ID in brackets


Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

How we do it

Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse

Elizabeth Manchen RN, MS, OCN,

, Caroline Robert MD, PhD and Camillo Porta MD

Received 12 August 2010; 

accepted 22 November 2010. 

Available online 13 February 2011.

Abstract

One significant toxicity associated with the anticancer tyrosine kinase inhibitors (TKIs) is hand–foot skin reaction (HFSR). We provide an overview of HFSR, emphasizing experience-based prevention techniques and nursing management strategies from the viewpoints of a medical oncologist, a dermatologist, and an oncology nurse. Supporting data include (1) published preclinical and phase I–III clinical studies and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib. HFSR has been reported in up to 60% of patients treated with sorafenib or sunitinib. TKI-induced HFSR may lead to dose reductions or treatment interruptions and reduced quality of life. Symptoms of TKI-associated HFSR can be managed by implementing supportive measures and aggressive dose modification. Patients educated about HFSR can work with their health-care teams to proactively detect and help manage this cutaneous toxicity, thus preventing or reducing the severity of TKI-associated HFSR. Successful prevention and management of TKI-associated HFSR can help to ensure that patients achieve optimal therapeutic outcomes. Implementation of such measures may increase the likelihood that therapy is continued for the appropriate interval at an appropriate dose for each patient. Optimal management of TKI-associated HFSR is predicated on establishing appropriate partnerships among medical oncologists, dermatologists, oncology nurses, and patients.

Article Outline

The Medical Oncologist's Viewpoint
Molecularly Targeted Agents
Characteristics of Hand–Foot Skin Reaction
Grading HFSR

The Dermatologist's Viewpoint
Incidence and Severity of HFSR With TKI
Management Strategies

The Oncology Nurse's Viewpoint

Summary

Acknowledgements

References

We are living in an era of “molecularly targeted therapy.” This targeted approach has developed as advances in science have led to a more detailed understanding of the inner workings of the cell, both in health and in illness. Once a molecular pathway has been implicated in the development and progression of cancer, modulators can be developed to intervene in this pathway. The goal is to target the cells and pathways specifically involved in the disease process, thereby leaving the normal processes undisturbed. The era of targeted therapy began in the late 1990s when the first targeted anticancer agents received regulatory approval (Figure 1). The targeted therapy armamentarium has since grown, creating a therapeutic landscape in which the chemical destruction of tumors with chemotoxic agents has been either expanded upon or replaced with agents designed to target carcinogenic processes.



Figure 1. 

Progress in the Medical Treatment of Cancer

This article reviews the mechanism of action, clinical trial results, and adverse effects of two molecularly targeted anticancer agents, the tyrosine kinase inhibitors (TKIs) sorafenib (Nexavar®; Bayer HealthCare Pharmaceuticals, Montville, NJ, and Onyx Pharmaceuticals, Emeryville, CA) and sunitinib (Sutent®; Pfizer Pharmaceuticals, New York, NY). This article specifically focuses on the diagnosis and management of TKI-associated hand–foot skin reaction (HFSR) from the perspectives of the medical oncologist, the dermatologist, and the oncology nurse. Data were derived from (1) published reports of preclinical and phase I–III clinical studies of sorafenib and sunitinib and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib.

The Medical Oncologist's Viewpoint

Molecularly Targeted Agents

Molecularly targeted therapies are directed at specific mechanisms involved in cell division, invasion, and metastasis, as well as in cell survival mediated by avoidance of apoptosis and resistance to conventional treatments. Clinical trials in several cancer types have shown that these TKIs can inhibit these activities of cancer cells by either cytostatic or cytotoxic mechanisms.1 However, the ability of these agents to inhibit multiple cancer cell pathways via novel mechanisms of action may explain, at least in part, their apparent direct toxic effects.2 These include adverse events that, from a medical viewpoint, must be anticipated, promptly recognized, and properly treated. Doing so can help minimize disruption to the patient's quality of life and may reduce the need for dose reduction or treatment interruption.1

Both sorafenib and sunitinib are orally administered, small-molecule inhibitors of multiple kinases, some of which are common to both agents (Figure 2).3 Sorafenib has known effects on tumor-cell proliferation and angiogenesis. Its antiproliferative effects are exerted via inhibition of serine/threonine kinases of the RAF/MEK/ERK signaling pathway (also called the MAP-kinase pathway) that is found within tumor cells; specifically, sorafenib targets wild-type RAF gene products (CRAF, BRAF) and mutant BRAF. The antiangiogenic effects of sorafenib are exerted via its inhibition of extracellular vascular endothelial growth factor (VEGF) receptors 2 and 3 (VEGFR-2 and VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-β), which is found mainly in the tumor vasculature. Sorafenib also exerts broad-spectrum activity against the stem-cell growth factor receptor (c-KIT), FMS-like tyrosine kinase 3 (Flt3), and the receptor encoded by the ret proto-oncogene (RET).[4], [5], [6] and [7] Sunitinib has demonstrated effects on the growth, pathologic angiogenesis, and metastatic progression of cancer by inhibiting PDGFR-α and -β; VEGFR-1, -2, and -3; and colony-stimulating factor receptor (CSF-1R), c-KIT, Flt3, and RET.8

 

 



Figure 2. 

Mechanisms of Action of Sorafenib and Sunitinib

Sorafenib and sunitinib specifically recognize and inhibit c-KIT, VEGFR, PDGFR-β, and Flt3 receptor tyrosine kinases. Sorafenib also inhibits RAF, a serine/threonine kinase involved in the RAF/MEK/ERK kinase pathway

c-KIT = stem-cell growth factor receptor (a cytokine receptor expressed on the surface of hematopoietic stem cells as well as other cell types); ERK = extracellular signal–regulated kinase; Flt3 = FMS-like tyrosine kinase 3 (a cytokine receptor expressed on the surface of hematopoietic progenitor cells); Flt3L = FMS-like tyrosine kinase 3 ligand (Flt3 ligand); MEK = (MAPK/ERK) kinase; PDGF-ΒΒ = platelet-derived growth factor BB; PDGFR-β = platelet-derived growth factor receptor beta; RAF = a gene that encodes for a protein kinase (Raf1) that functions in the mitogen-activated protein kinase/extracellular signal–regulated kinase (MAPK/ERK) signal-transduction pathway as part of a protein kinase cascade; RAS = a superfamily of genes that encode small GTPases involved in cellular signal transduction; SCF = stem-cell factor; VEGF = vascular endothelial growth factor; VEGFR = vascular endothelial growth factor receptor

Adapted with permission from Lacouture et al3

Sorafenib was approved for the treatment of advanced renal cell carcinoma (RCC) in 2005 and for unresectable hepatocellular carcinoma (HCC) in 2007. The efficacy of sorafenib in 903 patients with advanced RCC was demonstrated in the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial (TARGET), the largest phase III trial ever conducted in the second-line setting in patients with advanced RCC. Sorafenib significantly enhanced median progression-free survival (PFS) compared with placebo (24 vs 12 weeks),9 which led to early termination of the study and crossover from placebo to active drug. A preplanned analysis, which did not include patients who received placebo (who had crossed over to active treatment), ultimately demonstrated that sorafenib significantly prolonged overall survival (OS).10 Furthermore, 84% of sorafenib-treated patients experienced a clinical benefit, defined as objective response or disease stabilization.9 These results have been confirmed in larger, “real-world” patient populations in expanded-access programs conducted in North America (n = 2504)11 and the European Union (n = 118).12

Definitive data supporting the efficacy of sorafenib in HCC were provided by the randomized, double-blind, placebo-controlled Sorafenib CCC Assessment Randomized Protocol (SHARP) trial, the largest phase III trial ever conducted in patients with advanced HCC (n = 599) and the first phase III study to demonstrate a significant survival advantage with a systemic treatment in advanced HCC. In this trial, patients treated with sorafenib experienced a 44% increase in median OS (10.7 vs 7.9 months, hazard ratio [HR] = 0.69, 95% confidence interval [CI] 0.55–0.87, P < 0.001) and a 73% prolongation in median time to radiographic progression (5.5 vs 2.8 months, P < 0.001) compared with patients who received placebo.13 These results were confirmed in a separate phase III, randomized, double-blind, placebo-controlled study conducted in 226 patients from the Asia-Pacific region with advanced HCC.14 In this trial also, sorafenib significantly prolonged median OS (6.5 vs 4.2 months, HR = 0.68, 95% CI 0.50–0.93, P = 0.014) and time to progression (TTP) (2.8 vs 1.4 months, HR = 0.57, 95% CI 0.42–0.79, P = 0.0005) compared with placebo.

Sunitinib received approval in 2006 for use in patients with gastrointestinal stromal tumor (GIST) whose disease is refractory to imatinib (Gleevec®; Novartis Pharmaceuticals, East Hanover, NJ) or who are intolerant to the drug and in those with advanced RCC. Approval of sunitinib for the treatment of GIST was based on data from a randomized, placebo-controlled, phase III trial of 312 patients with imatinib-refractory GIST.15 In that study, sunitinib treatment increased median PFS (24.1 vs 6.0 weeks, HR = 0.33, 95% CI 0.24–0.47, P < 0.0001) and median TTP (27.3 vs 6.4 weeks, HR = 0.33, 95% CI 0.23–0.47, P < 0.0001) compared with placebo. The trial was unblinded early when a planned interim analysis revealed significantly longer TTP with sunitinib than with placebo. A subsequent analysis showed that median OS with sunitinib was about twice that with placebo (73.9 vs 35.7 weeks, P < 0.001).16 In an ongoing, worldwide treatment-use program to provide expanded access to sunitinib for patients with advanced GIST intolerant of or resistant to imatinib,17 sunitinib treatment resulted in a median estimated TTP of 41 weeks and a median estimated OS of 75 weeks in the population analyzed (n = 1,117).

A separate phase III randomized controlled trial was conducted in 750 patients with advanced RCC and no history of systemic therapy for RCC.[18] and [19] The active comparator in this trial was interferon-alfa (IFN-α). Compared with IFN-α, sunitinib significantly increased median PFS (11 vs 5 months, HR = 0.539, 95% CI 0.451–0.643, P < 0.001) and was associated with a greater objective response rate (47% vs 12%, P < 0.001). Median OS was greater in the sunitinib group (26.4 vs 21.8 months), but the difference was not significant (P = 0.051). Data from expanded-access programs in patients with RCC and GIST support the phase III trial data for sunitinib.20 Efficacy data for sorafenib and sunitinib are summarized in Table 1.[9], [10], [11], [12], [13], [14], [15], [17], [18] and [20]

 

 

Table 1. Summary of Efficacy Data for Sorafenib and Sunitinib

DRUG, DISEASE, AND STUDYEFFICACY DATA
Sorafenib
Advanced RCC
Phase III TARGET[9] and [10]

• Largest phase III trial ever conducted in second-line setting in patients with advanced RCC

• Median PFS was 5.5 months in sorafenib group vs 2.8 months in placebo group (P < 0.001)

• 28% improvement in OS vs placebo (HR = 0.72, P = 0.02)

• Significant prolongation of OS (HR = 0.78, 95% CI 0.62–0.97, P = 0.029)

• Clinical benefit (CR + PR + SD) in 84% of patients

Expanded-access programs[11] and [12]Data from expanded-access programs in community-based populations (NA- and EU-ARCCS) were consistent with data from TARGET
Unresectable HCC
Phase III SHARP trial13

• First phase III trial to demonstrate a significant survival advantage for a systemic therapy in advanced HCC

• Median OS was 10.7 months in sorafenib group vs 7.9 months in placebo group (HR = 0.69, 95% CI 0.55–0.87, P < 0.001)

• Median TTRP was 5.5 months in sorafenib group vs 2.8 months in placebo group (HR = 0.58, 95% CI 0.45–0.74, P < 0.001)

• Disease control rate (CR + PR + SD) was 43% in sorafenib group vs 32% in placebo group (P = 0.002)

Phase III Asia-Pacific trial14

• Median OS was 6.5 months in sorafenib group vs 4.2 months in placebo group (HR = 0.68, 95% CI 0.50–0.93, P = 0.014)

• Median TTP was 2.8 months in sorafenib group vs 1.4 months in placebo group (HR = 0.57, 95% CI 0.42–0.79, P = 0.0005)

Sunitinib
Advanced RCC
Phase III registration trial18

• Median PFS was 11 months in sunitinib group vs 5 months in interferon-α group (HR = 0.539, 95% CI 0.451–0.643, P < 0.001)

• Objective response rate (CR + PR) was 47% in sunitinib group vs 12% in interferon-α group (P < 0.001)

• Median OS was 26.4 months in sunitinib group vs 21.8 months in interferon-α group (P = 0.051)

Expanded-access program20

• In a broad population of patients with metastatic RCC who were treated with sunitinib:

 – Median PFS was 10.9 months

 – Median OS was 18.4 months

Imatinib-resistant GIST
Phase III registration trial15

• Median TTP was 27.3 weeks in sunitinib group vs 6.4 weeks in placebo group (HR = 0.33, 95% CI 0.23–0.47, P < 0.0001)

• Median PFS was 24.1 weeks in sunitinib group vs 6.0 weeks in placebo group (HR = 0.33, 95% CI 0.24–0.47, P < 0.0001)

• 16% of sunitinib-treated patients were progression-free for at least 26 weeks compared with 1% of those who received placebo

Expanded-access program17

• In a broad population of patients with imatinib-resistant GIST who were treated with sunitinib:

 – Estimated median TTP was 41 weeks

 – Estimated median OS was 75 weeks

CI = confidence interval; CR = complete response; EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; HR = hazard ratio; NA-ARCCS = North American ARCCS; OS = overall survival; PFS = progression-free survival; PR = partial response; RCC = renal cell carcinoma; SD = stable disease; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial; TTP = time to progression; TTRP = time to radiologic progression


Characteristics of Hand–Foot Skin Reaction

Data from the clinical trials for sorafenib and sunitinib indicate that both agents are generally well-tolerated; common treatment-related adverse reactions include diarrhea, alopecia, nausea, fatigue, rash, and hypertension, as well as palmar–plantar erythrodysesthesia (PPE) syndrome, also known as hand–foot skin reaction (HFSR) (Table 2).[10] and [19] HFSR is a dermatologic toxicity that has been reported in 14%–62% of patients treated with sorafenib or sunitinib (Table 3).[9], [11], [12], [13], [14], [15], [17], [18], [20], [21], [22], [23], [24] and [25] In general, the term HFSR refers to a group of signs and symptoms affecting the hands and feet of patients taking sorafenib, sunitinib, or, to a lesser extent, other TKIs such as pazopanib (Votrient™; GlaxoSmithKline, Research Triangle Park, NC)[26] and [27] and axitinib (AG013736).[28], [29], [30] and [31]

Table 2. Selected Common Adverse Events in Patients Treated with Sorafenib (n = 452) or Sunitinib (n = 375) in Phase III Registration Trials (Updated and Final Results)[10] and [19]

ADVERSE EVENT
SORAFENIB 400 MG BID
SUNITINIB 50 MG QD
ALL GRADES (%)GRADE 3/4 (%)ALL GRADES (%)GRADE 3/4 (%)
Diarrhea483619
Rash411242
Hand–foot skin reaction336299
Alopecia310120
Fatigue2935411
Nausea19<1525
Hypertension1743012
Dry skin13021<1
Vomiting121314
Mucositis50262

Table 3. Rates of Hand–Foot Skin Reaction in Clinical Trials of Sorafenib and Sunitinib

REFERENCESTUDYALL GRADES (%)GRADE 3 (%)GRADE 4 (%)
Sorafenib
 9Phase III TARGETa306 (grade 3/4)
 13Phase III SHARPa2180
 11NA-ARCCS, first-linea19 (≥2)11 (grade 3/4)
 11NA-ARCCS, second-linec17 (≥2)8 (grade 3/4)
 12EU-ARCCSa4712 (grade 3/4)
 14Phase III Asia-Pacifica4511 (grade 3/4)
 24Phase II randomized discontinuation trial in advanced RCCb62130
 25Phase II study in advanced HCCb3150
 21Phase II, uncontrolled study in relapsed/refractory NSCLCa3710 (grade 3/4)
Sunitinib
 18Phase III registration trial in advanced RCCa2050
 20Expanded access program in advanced RCCc5 (grade 3/4)
 15Phase III registration trial in imatinib-resistant GISTa1440
 [17] and [22]Expanded access program in imatinib-resistant GISTcN/A8 (grade 3/4)
 23Phase II trial of second-line treatment in advanced RCCa1570

EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; N/A = data not available; NA-ARCCS = North American ARCCS; NSCLC = non-small-cell lung cancer; RCC = renal cell carcinoma; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial

a Used version 3.0 of National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE)b Used version 2.0 of NCI-CTCAEc Version of NCI-CTCAE used not specified

 

 

HFSR is typically characterized by redness, marked discomfort, swelling, and tingling in the palms of the hands and/or soles of the feet.32 HFSR can be painful enough to interfere profoundly with activities of daily living (ADLs). In fact, patients may report symptoms after as few as 2 weeks on TKI therapy, at which point they may present to the health-care provider (HCP) wearing slippers, unable to walk, and having difficulty in performing ADLs such as eating, dressing, and bathing.[1] and [33] Although HFSR can lead to TKI dose modification or treatment discontinuation, preventive measures can be taken before TKIs are initiated to reduce the likelihood of HFSR. In addition, early treatment of symptoms may prevent HFSR from progressing to the point at which the patient's ability to receive the full potential benefit of therapy is compromised.[3], [34], [35] and [36]

Signs and symptoms of HFSR may appear concomitantly or sequentially and can affect both hands and both feet. Although symptoms are most prominent on the palms and soles, other areas of the hands and feet may also be involved, including the tips of the fingers and toes, the heels, and metatarsophalangeal skin; areas of flexure; and skin overlying the metacarpophalangeal and interphalangeal joints.3 These “pressure areas” are where the most severe symptoms are typically seen. Common symptoms include dysesthesia and paresthesia, described as “tingling, prickling, or creeping sensations” and/or sensitivity or intolerance to hot or warm objects (which may occur before other symptoms are apparent); erythema; edema; hyperkeratosis; and dry and/or cracked skin.[1] and [34] Actual HFSR lesions are described as tender and scaling, with a peripheral halo of erythema, yellowish and hyperkeratotic plaques, or callous-like blisters (which usually do not contain fluid), typically localized to areas of pressure.[3] and [35] Desquamation, particularly with sunitinib treatment, may also be present.37

Since both sorafenib and sunitinib inhibit the VEGFRs, PDGFRs, c-KIT, and Flt3,38 it is likely that inhibition of one or more of these receptors and/or pathways plays a role in HFSR development.36 Differences in the relative appearance of HFSR symptoms are dependent on whether sorafenib or sunitinib is used. Sunitinib use is more often associated with desquamation, whereas sorafenib is more often associated with areas of hyperkeratosis, particularly formation of thick calluses on the soles of the feet.37 The timing of the first appearance of symptoms may also vary according to the TKI used. HFSR usually develops within the first 2–4 weeks of treatment with a TKI and almost always within the first 6 weeks.35 However, because the severity of HFSR appears to be dose-dependent,3 signs and symptoms may present later rather than sooner in patients treated with sunitinib. This is likely due to the recommended sunitinib dosing schedule, which incorporates a 2-week period during which no drug is administered. Although HFSR frequently decreases in intensity during treatment, even without dose modifications or treatment interruption, prompt treatment of HFSR is recommended to prevent rapid progression. Early symptoms can usually be resolved easily by appropriate treatment, which often allows continuation of full-dose therapy for the prescribed length of time.

It is important to note what HFSR is not. TKI-associated HFSR is not the same clinical entity as the hand–foot syndrome (HFS) traditionally seen with cytotoxic agents such as infusional 5-fluorouracil (5-FU); capecitabine, the oral prodrug of 5-FU; and pegylated liposomal doxorubicin. Although HFSR and HFS share several clinical and pathological aspects—each previously has been called “acral erythema” and “PPE”—they clearly are not the same clinical or pathologic entity. HFSR is neither an allergic reaction to a drug nor an indication that a patient may be intolerant to a drug. Finally, HFSR does not indicate drug efficacy, as may be the case with skin rash in patients with non-small-cell lung cancer treated with erlotinib.[3] and [39]

Grading HFSR

In published reports, the severity of HFSR is usually graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE),32 a three-grade classification system. However, modified criteria are considered by some to be a better fit for routine clinical practice.1 What distinguishes the modified criteria from the NCI criteria (version 4.02) is the inclusion of HFSR-specific clinical characteristics plus certain patient-defined considerations used to categorize severity. The modified criteria expressly define the degree to which HFSR discomfort affects the patient's normal activities, an improvement over version 4.02 used alone. The NCI-CTCAE version 4.02 criteria, the modified criteria, and corresponding patient photographs are presented in Figure 3.[1] and [32]

 

 



Figure 3. 

National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE, version 4.02)32 and Modified Grading Criteria for Hand–Foot Skin Reaction1

Note that grade-4 toxicity is not contemplated in either system

Photographs reproduced with permission

Recommendations for the treatment of grade 1 HFSR include early and appropriate dermatologic management and active collaboration among HCPs.40

The Dermatologist's Viewpoint

Although the exact pathogenesis of HFSR has not been fully elucidated, research into its cause(s) is ongoing. Theoretically, traditional HFS is thought to be due to the direct toxic effects of drugs or their ability to invoke a “host-vs-host” response. In contrast, a unique mechanism has been proposed for TKI-associated HFSR: simultaneous blockage of VEGFRs and PDGFRs.2

Three histopathologic features have been found to predominate in HFSR: dyskeratotic keratinocytes at various states of necrosis (Figure 441), basal layer vacuolar degeneration, and mild perivascular or lichenoid lymphocyte-predominant infiltrate.2 Immunohistochemistry with a variety of skin-cell markers has shown a significant modification of normal maturation of keratinocytes, which are often apoptotic. Minor modifications of blood vessels are also seen, but no signs of intense vasculitis are evident. This is important because HFSR is suspected of being a “class effect” of TKIs that target VEGFRs. HFSR is not seen in patients treated with single-agent bevacizumab, and the lack of histologic evidence of significant damage to blood vessels suggests that HFSR does not result from the general inhibition of angiogenesis. A retrospective analysis found that HFSR rates were higher when patients were treated with sorafenib and bevacizumab in combination, supporting the hypothesis that HSFR is due to the anti-VEGF properties of sorafenib.42 Other possible causes of HFSR include activation by a ligand other than VEGF and/or inhibition of one of the other protein targets inhibited by both sorafenib and sunitinib.[3] and [35]



Figure 4. 

Layers of the Epidermis

The epidermis is composed of a very sophisticated arrangement of keratinocytes, which originate as stem cells in the stratum germinatum (not shown). The stem cells constantly multiply, creating daughter cells that progressively mature over approximately 28 days and move to the surface of the epidermis. As they move, they change their function and shape. In the stratum spinosum, interactions between cells resemble spines; mature cells move through the stratum granulosum, which has a very important secretory function, until finally the cells reach the stratum corneum and die. Thus, the stratum corneum is the layer of skin containing dead skin cells that have lost their nuclei; it is the part of the epidermis that ensures the barrier function of skin and is the layer most affected by HFSR. The layer beneath the epidermis is called the “papillary dermis.” The papillary dermis contains nerves and blood vessels and supplies the epidermis with nutrients. The fibroblasts and fibers located here give skin its strength and resistance

Adapted with permission from Gawkrodger41


Incidence and Severity of HFSR With TKI

To determine the incidence and severity of HFSR specific to sorafenib, a double-blind, prospective, dermatologic substudy was performed in patients enrolled in the phase III TARGET trial.35 Eighty-five patients with RCC were randomized to receive either sorafenib (n = 43) or placebo (n = 42). Dermatologic examinations were performed before and during treatment. Ninety-one percent of sorafenib-treated patients experienced at least one cutaneous reaction compared with 7% of those in the placebo group. A variant of HFSR clinically distinct from chemotherapy-induced HFS was observed in 60% of sorafenib-treated patients. Reversible grade 3 HFSR leading to dose reduction occurred in two sorafenib-treated patients. Additional cutaneous reactions were facial erythema, scalp dysesthesia, alopecia, and subungual splinter hemorrhages.

HFSR (of any grade) has been shown to occur in approximately 30% of patients treated with sorafenib and 20% of those who received sunitinib in clinical studies.43 Grade 3/4 HFSR has been observed in approximately 6% of sorafenib-treated and 5% of sunitinib-treated patients. HFSR was not reported in a phase II study of 142 patients with relapsed or refractory soft-tissue sarcoma treated with pazopanib.44 In a phase III randomized, double-blind, placebo-controlled trial of pazopanib in patients with advanced RCC, the incidence of HFSR was <10%, while the incidence of grade 3/4 HFSR was <1%. Potential differences may be explained by variations in the potency and selectivity of the TKIs.27

Management Strategies

Our work at the Dermatology Center at the Gustave-Roussy Institute has shown that early intervention against the dermatologic adverse effects of these TKIs can inhibit patient progression to a more serious form of HFSR.[34] and [38]

Effective management of HFSR can begin prior to initiation of treatment with sorafenib or sunitinib. Patients should be advised to remove any preexisting hyperkeratotic areas or calluses, keep skin well-moisturized with appropriate creams, and cushion pressure points with cotton socks, soft shoes, and/or insoles. Dose modification is typically not required for grade 1 HFSR; symptomatic treatments should be employed instead.

If HFSR symptoms progress to grade 2 or 3, with pain and a decrease in quality of life, the dose of sorafenib or sunitinib can be modified until symptoms recede, after which the patient can be brought back to the full dose. Very often, the patient can tolerate the full-dose treatment simply by decreasing the dose briefly.3 A recommended dose-modification scheme is shown in Figure 5.3

 

 



Figure 5. 

Management of Hand–Foot Skin Reaction by Grade

MKI = multikinase inhibitor

Adapted with permission from Lacouture et al3

TKIs are being studied in patients with additional types of tumor, possibly in the adjuvant setting, as well as in combinations. Because these drugs are administered orally, with a decreased (compared with conventional cytotoxic agents) frequency of nurse– or doctor–patient interactions, patients must be very well-informed of any potential toxicities with the TKIs.

At present, there are no evidence-based treatment guidelines for the prevention or management of HFSR. However, HCPs most involved in the day-to-day care of patients with HFSR have made great progress in establishing preventive and treatment strategies and in identifying ancillary products likely to decrease the incidence and/or severity of symptoms. Prevention, which includes preventing HFSR entirely as well as preventing progression from its initial appearance, is a key component of HFSR management.

The Oncology Nurse's Viewpoint

The nurse's viewpoint begins with patient education and empowerment. The goal is to prevent adverse effects from occurring while managing any adverse effects that do occur so that the patient has the best chance of staying on anticancer therapy. This requires a strong partnership between the HCP team and the patient. Although not all cases of HFSR can be prevented, experience suggests that symptom incidence and severity can be alleviated by educating patients to recognize the signs and symptoms of HFSR and report these to their HCPs. HFSR typically occurs early in the course of therapy, so it is prudent to be especially vigilant during the first 6 weeks. Providing the patient with a brochure about HFSR to refer to at home may facilitate identification of HFSR.

To address the lack of evidence-based guidelines to prevent or treat HFSR, an international, interdisciplinary expert panel has provided a set of consensus recommendations for the management of TKI-associated HFSR.45 One component of these recommendations can be phrased simply for the patients as a “3C” approach to management: control calluses, comfort with cushions, and cover with creams.

Prior to treatment, the patient should receive a full-body examination, with emphasis on the condition of the hands and feet. Evaluation should be performed by a qualified HCP who can determine whether there are physical conditions that may predispose a patient to areas of increased friction or rubbing. For all patients, especially those with comorbid conditions (eg, diabetes, poor circulation), a pretreatment pedicure is highly recommended. Patients should also be educated on the proper use of tools (eg, a pumice stone) to aid in callus removal. Such tools are considered beneficial because patients can control the frequency of their use and the extent of skin removed. However, because areas of hyperkeratosis are often extremely tender and painful, patients are cautioned against overuse of these tools, including the aggressive “paring” or “cutting” of callused areas. Finally, patients should be advised of the need for clean tools to guard against infection.

Other protective measures include the use of thick cotton gloves and/or socks, which may also help the skin to retain moisture, and avoidance of warm and/or hot water or objects, tight-fitting shoes, or other items that may rub, pinch, or cause friction in affected areas. Tender areas, pressure points, and pressure-sensitive areas of the hands and feet should be protected. For example, weight lifters might be advised to wear gloves. These recommendations hold true both before and after development of HFSR.[3], [33] and [35] Well-padded but nonconstrictive footwear should be worn, and the use of insole cushions or inserts (eg, silicone or gel) should be encouraged. Foot soaks with lukewarm water and magnesium sulfate may be soothing. Tender areas should be protected at all times, and patients should be encouraged not to walk barefoot.

Use of over-the-counter and prescription-strength creams and moisturizers during treatment with TKIs has also been recommended (Table 4).[40], [45], [46], [47], [48] and [49] Moisturizing agents should be applied liberally, immediately after bathing. Cotton gloves and/or socks can also be worn, to help retain moisture and to provide an additional layer of protection. When applied liberally, these products soften areas of thick and hardened skin, help keep the skin pliable, and may prevent cracks or breaks in skin integrity, which could cause additional discomfort. Prescription-strength topical agents have also shown anecdotal benefit (Table 4). These topical agents are typically applied twice daily to affected areas only because they may irritate unaffected skin. Data on the use of topical/systemic corticosteroids in the treatment of HFS remain inconclusive; the literature primarily includes case studies in patients with PPE treated with chemotherapeutic agents including pegylated liposomal doxorubicin.2 Finally, a qualified HCP must always be consulted to ensure proper diagnosis and treatment of HFSR.

 

 

Table 4. Selected Skin Care Products for Use in HFSR

Adapted with permission from Anderson et al40

PRODUCTSPRODUCT INFORMATION
Over-the-counter[40], [45] and [46]
 Cetaphil® (Galderma Laboratories, Ft. Worth, TX) skin cleaners, Aveeno® (Johnson & Johnson, New Brunswick, NJ) shower gelNondeodorant, fragrance-free products
 Udderly Smooth® (Redex Industries, Salem, OH), Gold Bond® (Chattem, Chattanooga, TN), Aveeno®Thicker products with more intense moisturizing properties than basic lotions; anti-itch formulations are available
 Norwegian Formula: Smoothing Relief Anti-Itch Moisturizer (Neutrogena, Los Angeles, CA)Contains dimethicone 1%, camphor 0.1%, and lidocaine
 Norwegian Formula: Foot Cream (Neutrogena)Contains cetearyl alcohol, dimethicone, menthol, and urea
 Bag Balm® (Dairy Association, Lyndonville, VT)May provide “cooling” effect from eucalyptus
 Eucerin® (Beiersdorf, Hamburg, Germany) CreamBest used at night due to greasy formulation
 Eucerin® Dry Skin TherapyContains urea and alpha-hydroxy acid
 Aquaphor® (Beiersdorf) Healing OintmentPetrolatum 41%
 Kerasal® (Alterna, Whippany, NJ)Salicylic acid 5% exfoliates and softens skin; urea 10% moisturizes skin
 Blue Lizard® (Crown Laboratories, Johnson City, TN)UV A and B sunblock, water-resistant
Prescription47–49
 Urea 40% cream (Carmol 40®; Doak Dermatologics, Fairfield, NJ)Softens hyperkeratosis, decreases epidermal thickness, proliferation
 Tazarotene 0.1% cream (Tazorac®; Allergan Dermatology, Irvine, CA)Retinoid decreases proliferation, reduces dermal inflammation
 Fluorouracil 5% cream (Carac®; Dermik Laboratories, Bridgewater, NJ)Antifolate inhibits proliferation; has shown anecdotal benefit in certain conditions characterized by hyperkeratotic lesions on the palms and soles


Summary

The addition of molecularly targeted agents to anticancer treatment has been found to cause both common and novel adverse reactions. HFSR is being increasingly recognized as a potential dose-limiting toxicity associated with sorafenib or sunitinib treatment that can result in discomfort, pain, decreased quality of life, and premature termination of a potentially effective cancer treatment. It is important to educate patients about potential dermatologic adverse effects associated with TKIs because limiting toxicity can help avoid treatment interruptions or dose reductions while improving ADLs.

The precise pathogenic mechanism of HFSR is currently not known, and there is no evidence-based protocol for treatment of HFSR. However, the increased clinical experience with these agents has resulted in a wealth of published articles describing empiric and symptomatic approaches that appear to help to prevent and manage HFSR. Frequent communication is necessary between the physician and patient, particularly 2–4 weeks from the initiation of therapy. Symptoms of HFSR should be recognized as early as possible. Providing the patient with a brochure about HFSR to refer to at home may facilitate the early identification of HFSR.

Patients should be advised of the “3C” approach to the management of TKI-associated HFSR: control calluses, comfort with cushions, and cover with creams. Creams should be applied after bathing and before going to bed; cotton gloves and socks should be worn to keep the cream on the hands and feet during the night.

Symptoms of HFSR typically are manageable with the implementation of supportive measures. If symptoms worsen, dose modification or interruption will result in a return to grade 0/1. Many patients can successfully be rechallenged with the full dose. Observations across multiple viewpoints have consistently shown that HFSR severity can be reduced in patients who are educated about HFSR and proactive about its detection and management.

Acknowledgments

All authors contributed equally to the development of this report. Editorial support was provided by Katherine Wright, PharmD, RPh, ISD, Wrighter Medical Education and Training, West Hills, CA; John A. Ibelli, CMPP, BelMed Professional Resources, New Rochelle, NY; and John D. Zoidis, MD, Bayer HealthCare Pharmaceuticals, Montville, NJ.

References1

1 C. Porta, C. Paglino, I. Imarisio and L. Bonomi, Uncovering Pandora's vase: the growing problem of new toxicities from novel anticancer agents: The case of sorafenib and sunitinib, Clin Exp Med 7 (2007), pp. 127–134. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

2 A.D. Lipworth, C. Robert and A.X. Zhu, Hand–foot syndrome (hand–foot skin reaction, palmar–plantar erythrodysesthesia): focus on sorafenib and sunitinib, Oncology 77 (2009), pp. 257–271 [19923864]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

3 M.E. Lacouture, S. Wu and C. Robert et al., Evolving strategies for the management of hand–foot skin reaction associated with the multitargeted kinase inhibitors sorafenib and sunitinib, Oncologist 13 (2008), pp. 1001–1011 [18779536]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (57)

4 S.M. Wilhelm, L. Adnane, P. Newell, A. Villanueva, J.M. Llovet and M. Lynch, Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling, Mol Cancer Ther 7 (2008), pp. 3129–3140 [18852116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)

5 D. Auclair, D. Miller and V. Yatsula et al., Antitumor activity of sorafenib in FLT3-driven leukemic cells, Leukemia 21 (2007), pp. 439–445 [17205056]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)

6 F. Carlomagno, S. Anaganti and T. Guida et al., BAY 43-9006 inhibition of oncogenic RET mutants, J Natl Cancer Inst 98 (2006), pp. 326–334 [16507829]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)

7 S.M. Wilhelm, C. Carter and L. Tang et al., BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis, Cancer Res 64 (2004), pp. 7099–7109 [15466206]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1142)

8 J.G. Christensen, A preclinical review of sunitinib, a multitargeted receptor tyrosine kinase inhibitor with anti-angiogenic and antitumour activities, Ann Oncol 10 (2007), pp. x3–x10 [17761721]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

9 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib in advanced clear-cell renal-cell carcinoma, N Engl J Med 356 (2007), pp. 125–134 [17215530]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1425)

10 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial, J Clin Oncol 27 (2009), pp. 3312–3318 [19451442]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (123)

11 W.M. Stadler, R.A. Figlin and D.F. McDermott et al., Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in North America, Cancer 116 (2010), pp. 1272–1280 [20082451]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)

12 J. Beck, E. Bajetta and B. Escudier et al., A large open-label, non-comparative, phase III study of the multi-targeted kinase inhibitor sorafenib in European patients with advanced renal cell carcinoma, Eur J Cancer (suppl) (2007), p. 5 Abstract 4506. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

13 J.M. Llovet, S. Ricci and V. Mazzaferro et al., Sorafenib in advanced hepatocellular carcinoma, N Engl J Med 359 (2008), pp. 378–390 [18650514]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (989)

14 A.L. Cheng, Y.K. Kang and Z. Chen et al., Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial, Lancet Oncol 10 (2009), pp. 25–34 [19095497]. Article |

PDF (195 K)
| View Record in Scopus | Cited By in Scopus (279)

15 G.D. Demetri, A.T. van Oosterom and C.R. Garrett et al., Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial, Lancet 368 (2006), pp. 1329–1338 [17046465]. Article |

PDF (200 K)
| View Record in Scopus | Cited By in Scopus (630)

16 G.D. Demetri, X. Huang and C.R. Garrett et al., Novel statistical analysis of long-term survival to account for crossover in a phase III trial of sunitinib (SU) vs placebo (PL) in advanced GIST after imatinib (IM) failure, J Clin Oncol (suppl) (2008), p. 26 Abstract 10524.

17 P. Reichardt, Y. Kang and W. Ruka et al., Detailed analysis of survival and safety with sunitinib (SU) in a worldwide treatment-use trial of patients with advanced GIST, J Clin Oncol (suppl) (2008), p. 26 Abstract 10548.

18 R.J. Motzer, T.E. Hutson and P. Tomczak et al., Sunitinib versus interferon alfa in metastatic renal-cell carcinoma, N Engl J Med 356 (2007), pp. 115–124 [17215529]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1520)

19R.J. Motzer, T.E. Hutson and P. Tomczak et al., Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 3584–3590 [19487381]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (194)

20 M.E. Gore, C. Szczylik and C. Porta et al., Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial, Lancet Oncol 10 (2009), pp. 757–763 [19615940]. Article |

PDF (142 K)
| View Record in Scopus | Cited By in Scopus (72)

21 G.R. Blumenschein Jr, U. Gatzemeier and F. Fossella et al., Phase II, multicenter, uncontrolled trial of single-agent sorafenib in patients with relapsed or refractory, advanced non-small-cell lung cancer, J Clin Oncol 27 (2009), pp. 4274–4280 [19652055]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

22 H.J. Schütte, M. Schlemmer, C. Wendtner, P. Reichardt and G.D. Demetri, Sunitinib (SU) in a worldwide treatment-use trial of pts with advanced gastrointestinal stromal tumours (GIST): analysis of survival and safety data, Onkologie (suppl 4) (2008), p. 201 Abstract V582.

23 R.J. Motzer, B.I. Rini and R.M. Bukowski et al., Sunitinib in patients with metastatic renal cell carcinoma, JAMA 295 (2006), pp. 2516–2524 [16757724]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (586)

24 M.J. Ratain, T. Eisen and W.M. Stadler et al., Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma, J Clin Oncol 24 (2006), pp. 2505–2512 [16636341]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (544)

25 G.K. Abou-Alfa, L. Schwartz and S. Ricci et al., Phase II study of sorafenib in patients with advanced hepatocellular carcinoma, J Clin Oncol 24 (2006), pp. 4293–4300 [16908937]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (418)

26 T.E. Hutson, I.D. Davis and J.P. Machiels et al., Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma, J Clin Oncol 28 (2010), pp. 475–480 [20008644]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

27 C.N. Sternberg, I.D. Davis and J. Mardiak et al., Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial, J Clin Oncol 28 (2010), pp. 1061–1068 [20100962]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (100)

28 O. Rixe, R.M. Bukowski and M.D. Michaelson et al., Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study, Lancet Oncol 8 (2007), pp. 975–984 [17959415]. Article |

PDF (143 K)
| View Record in Scopus | Cited By in Scopus (132)

29 B.I. Rini, I. Tamaskar and P. Shaheen et al., Hypothyroidism in patients with metastatic renal cell carcinoma treated with sunitinib, J Natl Cancer Inst 99 (2007), pp. 81–83 [17202116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (159)

30 B.I. Rini, G. Wilding and G. Hudes et al., Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 4462–4468 [19652060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (49)

31 H.S. Rugo, R.S. Herbst and G. Liu et al., Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: pharmacokinetic and clinical results, J Clin Oncol 23 (2005), pp. 5474–5483 [16027439]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (161)

32 National Cancer Institute, National Institutes of Health, US Department of Health and Human Services, Common Terminology Criteria for Adverse Events (CTCAE) Version 4.02. NIH publication 03-5410, National Institutes of Health, Bethesda, MD (2009).

33 L.S. Wood and B. Manchen, Sorafenib: a promising new targeted therapy for renal cell carcinoma, Clin J Oncol Nurs 11 (2007), pp. 649–656 [17962173]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)

34 C. Robert, C. Mateus, A. Spatz, J. Wechsler and B. Escudier, Dermatologic symptoms associated with the multikinase inhibitor sorafenib, J Am Acad Dermatol 60 (2009), pp. 299–305 [19028406]. Article |

PDF (296 K)
| View Record in Scopus | Cited By in Scopus (34)

35 J. Autier, B. Escudier, J. Wechsler, A. Spatz and C. Robert, Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor, Arch Dermatol 144 (2008), pp. 886–892 [18645140]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)

36 M.E. Lacouture, L.M. Reilly, P. Gerami and J. Guitart, Hand foot skin reaction in cancer patients treated with the multikinase inhibitors sorafenib and sunitinib, Ann Oncol 19 (2008), pp. 1955–1961 [18550575]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

37 L.S. Wood, Managing the side effects of sorafenib and sunitinib, Community Oncol 3 (2006), pp. 558–562. View Record in Scopus | Cited By in Scopus (19)

38 C. Robert, J.C. Soria and A. Spatz et al., Cutaneous side-effects of kinase inhibitors and blocking antibodies, Lancet Oncol 6 (2005), pp. 491–500 [15992698]. Article |

PDF (533 K)
| View Record in Scopus | Cited By in Scopus (208)

39 M.W. Saif, I. Merikas, S. Tsimboukis and K. Syrigos, Erlotinib-induced skin rash: Pathogenesis, clinical significance and management in pancreatic cancer patients, JOP 9 (2008), pp. 267–274 [18469438]. View Record in Scopus | Cited By in Scopus (11)

40 R. Anderson, A. Jatoi, C. Robert, L.S. Wood, K.N. Keating and M.E. Lacouture, Search for evidence-based approaches for the prevention and palliation of hand–foot skin reaction (HFSR) caused by the multikinase inhibitors (MKIs), Oncologist 14 (2009), pp. 291–302 [19276294]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)

41 D.J. Gawkrodger, Dermatology: An Illustrated Colour Text, 4th ed, Churchill Livingstone Elsevier, Edinburgh (2008).

42 N.S. Azad, J.B. Aragon-Ching and W.L. Dahut et al., Hand–foot skin reaction increases with cumulative sorafenib dose and with combination anti-vascular endothelial growth factor therapy, Clin Cancer Res 15 (2009), pp. 1411–1416 [19228742]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

43 N. Bhojani, C. Jeldres and J.J. Patard et al., Toxicities associated with the administration of sorafenib, sunitinib, and temsirolimus and their management in patients with metastatic renal cell carcinoma, Eur Urol 53 (2008), pp. 917–930 [18054825]. Article |

PDF (843 K)
| View Record in Scopus | Cited By in Scopus (81)

44 S. Sleijfer, I. Ray-Coquard and Z. Papai et al., Pazopanib, a multikinase angiogenesis inhibitor, in patients with relapsed or refractory advanced soft tissue sarcoma: a phase II study from the European Organisation for Research and Treatment of Cancer–Soft Tissue and Bone Sarcoma Group (EORTC study 62043), J Clin Oncol 27 (2009), pp. 3126–3132 [19451427]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)

45 L.S. Wood, H. Lemont and A. Jatoi et al., Practical considerations in the management of hand–foot skin reaction caused by multikinase inhibitors, Community Oncol 7 (2010), pp. 23–29. View Record in Scopus | Cited By in Scopus (3)

46 P. Esper, D. Gale and P. Muehlbauer, What kind of rash is it?: Deciphering the dermatologic toxicities of biologic and targeted therapies, Clin J Oncol Nurs 11 (2007), pp. 659–666 [17962174]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

47 I. Hagemann and E. Proksch, Topical treatment by urea reduces epidermal hyperproliferation and induces differentiation in psoriasis, Acta Derm Venereol 76 (1996), pp. 353–356 [8891006]. View Record in Scopus | Cited By in Scopus (41)

48 M. Duvic, A.T. Asano, C. Hager and S. Mays, The pathogenesis of psoriasis and the mechanism of action of tazarotene, J Am Acad Dermatol 39 (1998), pp. S129–S133 [9777790]. Article |

PDF (205 K)
| View Record in Scopus | Cited By in Scopus (24)

49 W. Scheithauer, J. McKendrick and S. Begbie et al., Oral capecitabine as an alternative to i.v. 5-fluorouracil-based adjuvant therapy for colon cancer: safety results of a randomized, phase III trial, Ann Oncol 14 (2003), pp. 1735–1743 [14630678]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (172)

 

 

Conflicts of interest: For E. M., none to disclose. C. R. has participated in advisory boards for Bayer, Roche, Pfizer, Astrazeneca, and GSK in the field of melanoma treatment and the management of the cutaneous side effects of anticancer agents. C. P. has acted as a paid adviser or speaker for Bayer Schering Pharma, Pfizer Oncology, Hoffman La Roche, Novartis Pharma, GSK, and Wyeth Pharmaceuticals and has received research funding from Bayer Schering Pharma and Novartis Pharma.

Correspondence to: Elizabeth Manchen, RN, MS, OCN, Section of Hematology/Oncology, University of Chicago Medical Center, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637; telephone: (773) 702–4135


1 PubMed ID in brackets


How we do it

Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse

Elizabeth Manchen RN, MS, OCN,

, Caroline Robert MD, PhD and Camillo Porta MD

Received 12 August 2010; 

accepted 22 November 2010. 

Available online 13 February 2011.

Abstract

One significant toxicity associated with the anticancer tyrosine kinase inhibitors (TKIs) is hand–foot skin reaction (HFSR). We provide an overview of HFSR, emphasizing experience-based prevention techniques and nursing management strategies from the viewpoints of a medical oncologist, a dermatologist, and an oncology nurse. Supporting data include (1) published preclinical and phase I–III clinical studies and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib. HFSR has been reported in up to 60% of patients treated with sorafenib or sunitinib. TKI-induced HFSR may lead to dose reductions or treatment interruptions and reduced quality of life. Symptoms of TKI-associated HFSR can be managed by implementing supportive measures and aggressive dose modification. Patients educated about HFSR can work with their health-care teams to proactively detect and help manage this cutaneous toxicity, thus preventing or reducing the severity of TKI-associated HFSR. Successful prevention and management of TKI-associated HFSR can help to ensure that patients achieve optimal therapeutic outcomes. Implementation of such measures may increase the likelihood that therapy is continued for the appropriate interval at an appropriate dose for each patient. Optimal management of TKI-associated HFSR is predicated on establishing appropriate partnerships among medical oncologists, dermatologists, oncology nurses, and patients.

Article Outline

The Medical Oncologist's Viewpoint
Molecularly Targeted Agents
Characteristics of Hand–Foot Skin Reaction
Grading HFSR

The Dermatologist's Viewpoint
Incidence and Severity of HFSR With TKI
Management Strategies

The Oncology Nurse's Viewpoint

Summary

Acknowledgements

References

We are living in an era of “molecularly targeted therapy.” This targeted approach has developed as advances in science have led to a more detailed understanding of the inner workings of the cell, both in health and in illness. Once a molecular pathway has been implicated in the development and progression of cancer, modulators can be developed to intervene in this pathway. The goal is to target the cells and pathways specifically involved in the disease process, thereby leaving the normal processes undisturbed. The era of targeted therapy began in the late 1990s when the first targeted anticancer agents received regulatory approval (Figure 1). The targeted therapy armamentarium has since grown, creating a therapeutic landscape in which the chemical destruction of tumors with chemotoxic agents has been either expanded upon or replaced with agents designed to target carcinogenic processes.



Figure 1. 

Progress in the Medical Treatment of Cancer

This article reviews the mechanism of action, clinical trial results, and adverse effects of two molecularly targeted anticancer agents, the tyrosine kinase inhibitors (TKIs) sorafenib (Nexavar®; Bayer HealthCare Pharmaceuticals, Montville, NJ, and Onyx Pharmaceuticals, Emeryville, CA) and sunitinib (Sutent®; Pfizer Pharmaceuticals, New York, NY). This article specifically focuses on the diagnosis and management of TKI-associated hand–foot skin reaction (HFSR) from the perspectives of the medical oncologist, the dermatologist, and the oncology nurse. Data were derived from (1) published reports of preclinical and phase I–III clinical studies of sorafenib and sunitinib and (2) published abstracts of phase II–III clinical trials of sorafenib and sunitinib.

The Medical Oncologist's Viewpoint

Molecularly Targeted Agents

Molecularly targeted therapies are directed at specific mechanisms involved in cell division, invasion, and metastasis, as well as in cell survival mediated by avoidance of apoptosis and resistance to conventional treatments. Clinical trials in several cancer types have shown that these TKIs can inhibit these activities of cancer cells by either cytostatic or cytotoxic mechanisms.1 However, the ability of these agents to inhibit multiple cancer cell pathways via novel mechanisms of action may explain, at least in part, their apparent direct toxic effects.2 These include adverse events that, from a medical viewpoint, must be anticipated, promptly recognized, and properly treated. Doing so can help minimize disruption to the patient's quality of life and may reduce the need for dose reduction or treatment interruption.1

Both sorafenib and sunitinib are orally administered, small-molecule inhibitors of multiple kinases, some of which are common to both agents (Figure 2).3 Sorafenib has known effects on tumor-cell proliferation and angiogenesis. Its antiproliferative effects are exerted via inhibition of serine/threonine kinases of the RAF/MEK/ERK signaling pathway (also called the MAP-kinase pathway) that is found within tumor cells; specifically, sorafenib targets wild-type RAF gene products (CRAF, BRAF) and mutant BRAF. The antiangiogenic effects of sorafenib are exerted via its inhibition of extracellular vascular endothelial growth factor (VEGF) receptors 2 and 3 (VEGFR-2 and VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-β), which is found mainly in the tumor vasculature. Sorafenib also exerts broad-spectrum activity against the stem-cell growth factor receptor (c-KIT), FMS-like tyrosine kinase 3 (Flt3), and the receptor encoded by the ret proto-oncogene (RET).[4], [5], [6] and [7] Sunitinib has demonstrated effects on the growth, pathologic angiogenesis, and metastatic progression of cancer by inhibiting PDGFR-α and -β; VEGFR-1, -2, and -3; and colony-stimulating factor receptor (CSF-1R), c-KIT, Flt3, and RET.8

 

 



Figure 2. 

Mechanisms of Action of Sorafenib and Sunitinib

Sorafenib and sunitinib specifically recognize and inhibit c-KIT, VEGFR, PDGFR-β, and Flt3 receptor tyrosine kinases. Sorafenib also inhibits RAF, a serine/threonine kinase involved in the RAF/MEK/ERK kinase pathway

c-KIT = stem-cell growth factor receptor (a cytokine receptor expressed on the surface of hematopoietic stem cells as well as other cell types); ERK = extracellular signal–regulated kinase; Flt3 = FMS-like tyrosine kinase 3 (a cytokine receptor expressed on the surface of hematopoietic progenitor cells); Flt3L = FMS-like tyrosine kinase 3 ligand (Flt3 ligand); MEK = (MAPK/ERK) kinase; PDGF-ΒΒ = platelet-derived growth factor BB; PDGFR-β = platelet-derived growth factor receptor beta; RAF = a gene that encodes for a protein kinase (Raf1) that functions in the mitogen-activated protein kinase/extracellular signal–regulated kinase (MAPK/ERK) signal-transduction pathway as part of a protein kinase cascade; RAS = a superfamily of genes that encode small GTPases involved in cellular signal transduction; SCF = stem-cell factor; VEGF = vascular endothelial growth factor; VEGFR = vascular endothelial growth factor receptor

Adapted with permission from Lacouture et al3

Sorafenib was approved for the treatment of advanced renal cell carcinoma (RCC) in 2005 and for unresectable hepatocellular carcinoma (HCC) in 2007. The efficacy of sorafenib in 903 patients with advanced RCC was demonstrated in the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial (TARGET), the largest phase III trial ever conducted in the second-line setting in patients with advanced RCC. Sorafenib significantly enhanced median progression-free survival (PFS) compared with placebo (24 vs 12 weeks),9 which led to early termination of the study and crossover from placebo to active drug. A preplanned analysis, which did not include patients who received placebo (who had crossed over to active treatment), ultimately demonstrated that sorafenib significantly prolonged overall survival (OS).10 Furthermore, 84% of sorafenib-treated patients experienced a clinical benefit, defined as objective response or disease stabilization.9 These results have been confirmed in larger, “real-world” patient populations in expanded-access programs conducted in North America (n = 2504)11 and the European Union (n = 118).12

Definitive data supporting the efficacy of sorafenib in HCC were provided by the randomized, double-blind, placebo-controlled Sorafenib CCC Assessment Randomized Protocol (SHARP) trial, the largest phase III trial ever conducted in patients with advanced HCC (n = 599) and the first phase III study to demonstrate a significant survival advantage with a systemic treatment in advanced HCC. In this trial, patients treated with sorafenib experienced a 44% increase in median OS (10.7 vs 7.9 months, hazard ratio [HR] = 0.69, 95% confidence interval [CI] 0.55–0.87, P < 0.001) and a 73% prolongation in median time to radiographic progression (5.5 vs 2.8 months, P < 0.001) compared with patients who received placebo.13 These results were confirmed in a separate phase III, randomized, double-blind, placebo-controlled study conducted in 226 patients from the Asia-Pacific region with advanced HCC.14 In this trial also, sorafenib significantly prolonged median OS (6.5 vs 4.2 months, HR = 0.68, 95% CI 0.50–0.93, P = 0.014) and time to progression (TTP) (2.8 vs 1.4 months, HR = 0.57, 95% CI 0.42–0.79, P = 0.0005) compared with placebo.

Sunitinib received approval in 2006 for use in patients with gastrointestinal stromal tumor (GIST) whose disease is refractory to imatinib (Gleevec®; Novartis Pharmaceuticals, East Hanover, NJ) or who are intolerant to the drug and in those with advanced RCC. Approval of sunitinib for the treatment of GIST was based on data from a randomized, placebo-controlled, phase III trial of 312 patients with imatinib-refractory GIST.15 In that study, sunitinib treatment increased median PFS (24.1 vs 6.0 weeks, HR = 0.33, 95% CI 0.24–0.47, P < 0.0001) and median TTP (27.3 vs 6.4 weeks, HR = 0.33, 95% CI 0.23–0.47, P < 0.0001) compared with placebo. The trial was unblinded early when a planned interim analysis revealed significantly longer TTP with sunitinib than with placebo. A subsequent analysis showed that median OS with sunitinib was about twice that with placebo (73.9 vs 35.7 weeks, P < 0.001).16 In an ongoing, worldwide treatment-use program to provide expanded access to sunitinib for patients with advanced GIST intolerant of or resistant to imatinib,17 sunitinib treatment resulted in a median estimated TTP of 41 weeks and a median estimated OS of 75 weeks in the population analyzed (n = 1,117).

A separate phase III randomized controlled trial was conducted in 750 patients with advanced RCC and no history of systemic therapy for RCC.[18] and [19] The active comparator in this trial was interferon-alfa (IFN-α). Compared with IFN-α, sunitinib significantly increased median PFS (11 vs 5 months, HR = 0.539, 95% CI 0.451–0.643, P < 0.001) and was associated with a greater objective response rate (47% vs 12%, P < 0.001). Median OS was greater in the sunitinib group (26.4 vs 21.8 months), but the difference was not significant (P = 0.051). Data from expanded-access programs in patients with RCC and GIST support the phase III trial data for sunitinib.20 Efficacy data for sorafenib and sunitinib are summarized in Table 1.[9], [10], [11], [12], [13], [14], [15], [17], [18] and [20]

 

 

Table 1. Summary of Efficacy Data for Sorafenib and Sunitinib

DRUG, DISEASE, AND STUDYEFFICACY DATA
Sorafenib
Advanced RCC
Phase III TARGET[9] and [10]

• Largest phase III trial ever conducted in second-line setting in patients with advanced RCC

• Median PFS was 5.5 months in sorafenib group vs 2.8 months in placebo group (P < 0.001)

• 28% improvement in OS vs placebo (HR = 0.72, P = 0.02)

• Significant prolongation of OS (HR = 0.78, 95% CI 0.62–0.97, P = 0.029)

• Clinical benefit (CR + PR + SD) in 84% of patients

Expanded-access programs[11] and [12]Data from expanded-access programs in community-based populations (NA- and EU-ARCCS) were consistent with data from TARGET
Unresectable HCC
Phase III SHARP trial13

• First phase III trial to demonstrate a significant survival advantage for a systemic therapy in advanced HCC

• Median OS was 10.7 months in sorafenib group vs 7.9 months in placebo group (HR = 0.69, 95% CI 0.55–0.87, P < 0.001)

• Median TTRP was 5.5 months in sorafenib group vs 2.8 months in placebo group (HR = 0.58, 95% CI 0.45–0.74, P < 0.001)

• Disease control rate (CR + PR + SD) was 43% in sorafenib group vs 32% in placebo group (P = 0.002)

Phase III Asia-Pacific trial14

• Median OS was 6.5 months in sorafenib group vs 4.2 months in placebo group (HR = 0.68, 95% CI 0.50–0.93, P = 0.014)

• Median TTP was 2.8 months in sorafenib group vs 1.4 months in placebo group (HR = 0.57, 95% CI 0.42–0.79, P = 0.0005)

Sunitinib
Advanced RCC
Phase III registration trial18

• Median PFS was 11 months in sunitinib group vs 5 months in interferon-α group (HR = 0.539, 95% CI 0.451–0.643, P < 0.001)

• Objective response rate (CR + PR) was 47% in sunitinib group vs 12% in interferon-α group (P < 0.001)

• Median OS was 26.4 months in sunitinib group vs 21.8 months in interferon-α group (P = 0.051)

Expanded-access program20

• In a broad population of patients with metastatic RCC who were treated with sunitinib:

 – Median PFS was 10.9 months

 – Median OS was 18.4 months

Imatinib-resistant GIST
Phase III registration trial15

• Median TTP was 27.3 weeks in sunitinib group vs 6.4 weeks in placebo group (HR = 0.33, 95% CI 0.23–0.47, P < 0.0001)

• Median PFS was 24.1 weeks in sunitinib group vs 6.0 weeks in placebo group (HR = 0.33, 95% CI 0.24–0.47, P < 0.0001)

• 16% of sunitinib-treated patients were progression-free for at least 26 weeks compared with 1% of those who received placebo

Expanded-access program17

• In a broad population of patients with imatinib-resistant GIST who were treated with sunitinib:

 – Estimated median TTP was 41 weeks

 – Estimated median OS was 75 weeks

CI = confidence interval; CR = complete response; EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; HR = hazard ratio; NA-ARCCS = North American ARCCS; OS = overall survival; PFS = progression-free survival; PR = partial response; RCC = renal cell carcinoma; SD = stable disease; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial; TTP = time to progression; TTRP = time to radiologic progression


Characteristics of Hand–Foot Skin Reaction

Data from the clinical trials for sorafenib and sunitinib indicate that both agents are generally well-tolerated; common treatment-related adverse reactions include diarrhea, alopecia, nausea, fatigue, rash, and hypertension, as well as palmar–plantar erythrodysesthesia (PPE) syndrome, also known as hand–foot skin reaction (HFSR) (Table 2).[10] and [19] HFSR is a dermatologic toxicity that has been reported in 14%–62% of patients treated with sorafenib or sunitinib (Table 3).[9], [11], [12], [13], [14], [15], [17], [18], [20], [21], [22], [23], [24] and [25] In general, the term HFSR refers to a group of signs and symptoms affecting the hands and feet of patients taking sorafenib, sunitinib, or, to a lesser extent, other TKIs such as pazopanib (Votrient™; GlaxoSmithKline, Research Triangle Park, NC)[26] and [27] and axitinib (AG013736).[28], [29], [30] and [31]

Table 2. Selected Common Adverse Events in Patients Treated with Sorafenib (n = 452) or Sunitinib (n = 375) in Phase III Registration Trials (Updated and Final Results)[10] and [19]

ADVERSE EVENT
SORAFENIB 400 MG BID
SUNITINIB 50 MG QD
ALL GRADES (%)GRADE 3/4 (%)ALL GRADES (%)GRADE 3/4 (%)
Diarrhea483619
Rash411242
Hand–foot skin reaction336299
Alopecia310120
Fatigue2935411
Nausea19<1525
Hypertension1743012
Dry skin13021<1
Vomiting121314
Mucositis50262

Table 3. Rates of Hand–Foot Skin Reaction in Clinical Trials of Sorafenib and Sunitinib

REFERENCESTUDYALL GRADES (%)GRADE 3 (%)GRADE 4 (%)
Sorafenib
 9Phase III TARGETa306 (grade 3/4)
 13Phase III SHARPa2180
 11NA-ARCCS, first-linea19 (≥2)11 (grade 3/4)
 11NA-ARCCS, second-linec17 (≥2)8 (grade 3/4)
 12EU-ARCCSa4712 (grade 3/4)
 14Phase III Asia-Pacifica4511 (grade 3/4)
 24Phase II randomized discontinuation trial in advanced RCCb62130
 25Phase II study in advanced HCCb3150
 21Phase II, uncontrolled study in relapsed/refractory NSCLCa3710 (grade 3/4)
Sunitinib
 18Phase III registration trial in advanced RCCa2050
 20Expanded access program in advanced RCCc5 (grade 3/4)
 15Phase III registration trial in imatinib-resistant GISTa1440
 [17] and [22]Expanded access program in imatinib-resistant GISTcN/A8 (grade 3/4)
 23Phase II trial of second-line treatment in advanced RCCa1570

EU-ARCCS = European Union Advanced Renal Cell Carcinoma Sorafenib; GIST = gastrointestinal stromal tumor; HCC = hepatocellular carcinoma; N/A = data not available; NA-ARCCS = North American ARCCS; NSCLC = non-small-cell lung cancer; RCC = renal cell carcinoma; SHARP = Sorafenib CCC Assessment Randomized Protocol; TARGET = Treatment Approaches in Renal Cancer Global Evaluation Trial

a Used version 3.0 of National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE)b Used version 2.0 of NCI-CTCAEc Version of NCI-CTCAE used not specified

 

 

HFSR is typically characterized by redness, marked discomfort, swelling, and tingling in the palms of the hands and/or soles of the feet.32 HFSR can be painful enough to interfere profoundly with activities of daily living (ADLs). In fact, patients may report symptoms after as few as 2 weeks on TKI therapy, at which point they may present to the health-care provider (HCP) wearing slippers, unable to walk, and having difficulty in performing ADLs such as eating, dressing, and bathing.[1] and [33] Although HFSR can lead to TKI dose modification or treatment discontinuation, preventive measures can be taken before TKIs are initiated to reduce the likelihood of HFSR. In addition, early treatment of symptoms may prevent HFSR from progressing to the point at which the patient's ability to receive the full potential benefit of therapy is compromised.[3], [34], [35] and [36]

Signs and symptoms of HFSR may appear concomitantly or sequentially and can affect both hands and both feet. Although symptoms are most prominent on the palms and soles, other areas of the hands and feet may also be involved, including the tips of the fingers and toes, the heels, and metatarsophalangeal skin; areas of flexure; and skin overlying the metacarpophalangeal and interphalangeal joints.3 These “pressure areas” are where the most severe symptoms are typically seen. Common symptoms include dysesthesia and paresthesia, described as “tingling, prickling, or creeping sensations” and/or sensitivity or intolerance to hot or warm objects (which may occur before other symptoms are apparent); erythema; edema; hyperkeratosis; and dry and/or cracked skin.[1] and [34] Actual HFSR lesions are described as tender and scaling, with a peripheral halo of erythema, yellowish and hyperkeratotic plaques, or callous-like blisters (which usually do not contain fluid), typically localized to areas of pressure.[3] and [35] Desquamation, particularly with sunitinib treatment, may also be present.37

Since both sorafenib and sunitinib inhibit the VEGFRs, PDGFRs, c-KIT, and Flt3,38 it is likely that inhibition of one or more of these receptors and/or pathways plays a role in HFSR development.36 Differences in the relative appearance of HFSR symptoms are dependent on whether sorafenib or sunitinib is used. Sunitinib use is more often associated with desquamation, whereas sorafenib is more often associated with areas of hyperkeratosis, particularly formation of thick calluses on the soles of the feet.37 The timing of the first appearance of symptoms may also vary according to the TKI used. HFSR usually develops within the first 2–4 weeks of treatment with a TKI and almost always within the first 6 weeks.35 However, because the severity of HFSR appears to be dose-dependent,3 signs and symptoms may present later rather than sooner in patients treated with sunitinib. This is likely due to the recommended sunitinib dosing schedule, which incorporates a 2-week period during which no drug is administered. Although HFSR frequently decreases in intensity during treatment, even without dose modifications or treatment interruption, prompt treatment of HFSR is recommended to prevent rapid progression. Early symptoms can usually be resolved easily by appropriate treatment, which often allows continuation of full-dose therapy for the prescribed length of time.

It is important to note what HFSR is not. TKI-associated HFSR is not the same clinical entity as the hand–foot syndrome (HFS) traditionally seen with cytotoxic agents such as infusional 5-fluorouracil (5-FU); capecitabine, the oral prodrug of 5-FU; and pegylated liposomal doxorubicin. Although HFSR and HFS share several clinical and pathological aspects—each previously has been called “acral erythema” and “PPE”—they clearly are not the same clinical or pathologic entity. HFSR is neither an allergic reaction to a drug nor an indication that a patient may be intolerant to a drug. Finally, HFSR does not indicate drug efficacy, as may be the case with skin rash in patients with non-small-cell lung cancer treated with erlotinib.[3] and [39]

Grading HFSR

In published reports, the severity of HFSR is usually graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE),32 a three-grade classification system. However, modified criteria are considered by some to be a better fit for routine clinical practice.1 What distinguishes the modified criteria from the NCI criteria (version 4.02) is the inclusion of HFSR-specific clinical characteristics plus certain patient-defined considerations used to categorize severity. The modified criteria expressly define the degree to which HFSR discomfort affects the patient's normal activities, an improvement over version 4.02 used alone. The NCI-CTCAE version 4.02 criteria, the modified criteria, and corresponding patient photographs are presented in Figure 3.[1] and [32]

 

 



Figure 3. 

National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE, version 4.02)32 and Modified Grading Criteria for Hand–Foot Skin Reaction1

Note that grade-4 toxicity is not contemplated in either system

Photographs reproduced with permission

Recommendations for the treatment of grade 1 HFSR include early and appropriate dermatologic management and active collaboration among HCPs.40

The Dermatologist's Viewpoint

Although the exact pathogenesis of HFSR has not been fully elucidated, research into its cause(s) is ongoing. Theoretically, traditional HFS is thought to be due to the direct toxic effects of drugs or their ability to invoke a “host-vs-host” response. In contrast, a unique mechanism has been proposed for TKI-associated HFSR: simultaneous blockage of VEGFRs and PDGFRs.2

Three histopathologic features have been found to predominate in HFSR: dyskeratotic keratinocytes at various states of necrosis (Figure 441), basal layer vacuolar degeneration, and mild perivascular or lichenoid lymphocyte-predominant infiltrate.2 Immunohistochemistry with a variety of skin-cell markers has shown a significant modification of normal maturation of keratinocytes, which are often apoptotic. Minor modifications of blood vessels are also seen, but no signs of intense vasculitis are evident. This is important because HFSR is suspected of being a “class effect” of TKIs that target VEGFRs. HFSR is not seen in patients treated with single-agent bevacizumab, and the lack of histologic evidence of significant damage to blood vessels suggests that HFSR does not result from the general inhibition of angiogenesis. A retrospective analysis found that HFSR rates were higher when patients were treated with sorafenib and bevacizumab in combination, supporting the hypothesis that HSFR is due to the anti-VEGF properties of sorafenib.42 Other possible causes of HFSR include activation by a ligand other than VEGF and/or inhibition of one of the other protein targets inhibited by both sorafenib and sunitinib.[3] and [35]



Figure 4. 

Layers of the Epidermis

The epidermis is composed of a very sophisticated arrangement of keratinocytes, which originate as stem cells in the stratum germinatum (not shown). The stem cells constantly multiply, creating daughter cells that progressively mature over approximately 28 days and move to the surface of the epidermis. As they move, they change their function and shape. In the stratum spinosum, interactions between cells resemble spines; mature cells move through the stratum granulosum, which has a very important secretory function, until finally the cells reach the stratum corneum and die. Thus, the stratum corneum is the layer of skin containing dead skin cells that have lost their nuclei; it is the part of the epidermis that ensures the barrier function of skin and is the layer most affected by HFSR. The layer beneath the epidermis is called the “papillary dermis.” The papillary dermis contains nerves and blood vessels and supplies the epidermis with nutrients. The fibroblasts and fibers located here give skin its strength and resistance

Adapted with permission from Gawkrodger41


Incidence and Severity of HFSR With TKI

To determine the incidence and severity of HFSR specific to sorafenib, a double-blind, prospective, dermatologic substudy was performed in patients enrolled in the phase III TARGET trial.35 Eighty-five patients with RCC were randomized to receive either sorafenib (n = 43) or placebo (n = 42). Dermatologic examinations were performed before and during treatment. Ninety-one percent of sorafenib-treated patients experienced at least one cutaneous reaction compared with 7% of those in the placebo group. A variant of HFSR clinically distinct from chemotherapy-induced HFS was observed in 60% of sorafenib-treated patients. Reversible grade 3 HFSR leading to dose reduction occurred in two sorafenib-treated patients. Additional cutaneous reactions were facial erythema, scalp dysesthesia, alopecia, and subungual splinter hemorrhages.

HFSR (of any grade) has been shown to occur in approximately 30% of patients treated with sorafenib and 20% of those who received sunitinib in clinical studies.43 Grade 3/4 HFSR has been observed in approximately 6% of sorafenib-treated and 5% of sunitinib-treated patients. HFSR was not reported in a phase II study of 142 patients with relapsed or refractory soft-tissue sarcoma treated with pazopanib.44 In a phase III randomized, double-blind, placebo-controlled trial of pazopanib in patients with advanced RCC, the incidence of HFSR was <10%, while the incidence of grade 3/4 HFSR was <1%. Potential differences may be explained by variations in the potency and selectivity of the TKIs.27

Management Strategies

Our work at the Dermatology Center at the Gustave-Roussy Institute has shown that early intervention against the dermatologic adverse effects of these TKIs can inhibit patient progression to a more serious form of HFSR.[34] and [38]

Effective management of HFSR can begin prior to initiation of treatment with sorafenib or sunitinib. Patients should be advised to remove any preexisting hyperkeratotic areas or calluses, keep skin well-moisturized with appropriate creams, and cushion pressure points with cotton socks, soft shoes, and/or insoles. Dose modification is typically not required for grade 1 HFSR; symptomatic treatments should be employed instead.

If HFSR symptoms progress to grade 2 or 3, with pain and a decrease in quality of life, the dose of sorafenib or sunitinib can be modified until symptoms recede, after which the patient can be brought back to the full dose. Very often, the patient can tolerate the full-dose treatment simply by decreasing the dose briefly.3 A recommended dose-modification scheme is shown in Figure 5.3

 

 



Figure 5. 

Management of Hand–Foot Skin Reaction by Grade

MKI = multikinase inhibitor

Adapted with permission from Lacouture et al3

TKIs are being studied in patients with additional types of tumor, possibly in the adjuvant setting, as well as in combinations. Because these drugs are administered orally, with a decreased (compared with conventional cytotoxic agents) frequency of nurse– or doctor–patient interactions, patients must be very well-informed of any potential toxicities with the TKIs.

At present, there are no evidence-based treatment guidelines for the prevention or management of HFSR. However, HCPs most involved in the day-to-day care of patients with HFSR have made great progress in establishing preventive and treatment strategies and in identifying ancillary products likely to decrease the incidence and/or severity of symptoms. Prevention, which includes preventing HFSR entirely as well as preventing progression from its initial appearance, is a key component of HFSR management.

The Oncology Nurse's Viewpoint

The nurse's viewpoint begins with patient education and empowerment. The goal is to prevent adverse effects from occurring while managing any adverse effects that do occur so that the patient has the best chance of staying on anticancer therapy. This requires a strong partnership between the HCP team and the patient. Although not all cases of HFSR can be prevented, experience suggests that symptom incidence and severity can be alleviated by educating patients to recognize the signs and symptoms of HFSR and report these to their HCPs. HFSR typically occurs early in the course of therapy, so it is prudent to be especially vigilant during the first 6 weeks. Providing the patient with a brochure about HFSR to refer to at home may facilitate identification of HFSR.

To address the lack of evidence-based guidelines to prevent or treat HFSR, an international, interdisciplinary expert panel has provided a set of consensus recommendations for the management of TKI-associated HFSR.45 One component of these recommendations can be phrased simply for the patients as a “3C” approach to management: control calluses, comfort with cushions, and cover with creams.

Prior to treatment, the patient should receive a full-body examination, with emphasis on the condition of the hands and feet. Evaluation should be performed by a qualified HCP who can determine whether there are physical conditions that may predispose a patient to areas of increased friction or rubbing. For all patients, especially those with comorbid conditions (eg, diabetes, poor circulation), a pretreatment pedicure is highly recommended. Patients should also be educated on the proper use of tools (eg, a pumice stone) to aid in callus removal. Such tools are considered beneficial because patients can control the frequency of their use and the extent of skin removed. However, because areas of hyperkeratosis are often extremely tender and painful, patients are cautioned against overuse of these tools, including the aggressive “paring” or “cutting” of callused areas. Finally, patients should be advised of the need for clean tools to guard against infection.

Other protective measures include the use of thick cotton gloves and/or socks, which may also help the skin to retain moisture, and avoidance of warm and/or hot water or objects, tight-fitting shoes, or other items that may rub, pinch, or cause friction in affected areas. Tender areas, pressure points, and pressure-sensitive areas of the hands and feet should be protected. For example, weight lifters might be advised to wear gloves. These recommendations hold true both before and after development of HFSR.[3], [33] and [35] Well-padded but nonconstrictive footwear should be worn, and the use of insole cushions or inserts (eg, silicone or gel) should be encouraged. Foot soaks with lukewarm water and magnesium sulfate may be soothing. Tender areas should be protected at all times, and patients should be encouraged not to walk barefoot.

Use of over-the-counter and prescription-strength creams and moisturizers during treatment with TKIs has also been recommended (Table 4).[40], [45], [46], [47], [48] and [49] Moisturizing agents should be applied liberally, immediately after bathing. Cotton gloves and/or socks can also be worn, to help retain moisture and to provide an additional layer of protection. When applied liberally, these products soften areas of thick and hardened skin, help keep the skin pliable, and may prevent cracks or breaks in skin integrity, which could cause additional discomfort. Prescription-strength topical agents have also shown anecdotal benefit (Table 4). These topical agents are typically applied twice daily to affected areas only because they may irritate unaffected skin. Data on the use of topical/systemic corticosteroids in the treatment of HFS remain inconclusive; the literature primarily includes case studies in patients with PPE treated with chemotherapeutic agents including pegylated liposomal doxorubicin.2 Finally, a qualified HCP must always be consulted to ensure proper diagnosis and treatment of HFSR.

 

 

Table 4. Selected Skin Care Products for Use in HFSR

Adapted with permission from Anderson et al40

PRODUCTSPRODUCT INFORMATION
Over-the-counter[40], [45] and [46]
 Cetaphil® (Galderma Laboratories, Ft. Worth, TX) skin cleaners, Aveeno® (Johnson & Johnson, New Brunswick, NJ) shower gelNondeodorant, fragrance-free products
 Udderly Smooth® (Redex Industries, Salem, OH), Gold Bond® (Chattem, Chattanooga, TN), Aveeno®Thicker products with more intense moisturizing properties than basic lotions; anti-itch formulations are available
 Norwegian Formula: Smoothing Relief Anti-Itch Moisturizer (Neutrogena, Los Angeles, CA)Contains dimethicone 1%, camphor 0.1%, and lidocaine
 Norwegian Formula: Foot Cream (Neutrogena)Contains cetearyl alcohol, dimethicone, menthol, and urea
 Bag Balm® (Dairy Association, Lyndonville, VT)May provide “cooling” effect from eucalyptus
 Eucerin® (Beiersdorf, Hamburg, Germany) CreamBest used at night due to greasy formulation
 Eucerin® Dry Skin TherapyContains urea and alpha-hydroxy acid
 Aquaphor® (Beiersdorf) Healing OintmentPetrolatum 41%
 Kerasal® (Alterna, Whippany, NJ)Salicylic acid 5% exfoliates and softens skin; urea 10% moisturizes skin
 Blue Lizard® (Crown Laboratories, Johnson City, TN)UV A and B sunblock, water-resistant
Prescription47–49
 Urea 40% cream (Carmol 40®; Doak Dermatologics, Fairfield, NJ)Softens hyperkeratosis, decreases epidermal thickness, proliferation
 Tazarotene 0.1% cream (Tazorac®; Allergan Dermatology, Irvine, CA)Retinoid decreases proliferation, reduces dermal inflammation
 Fluorouracil 5% cream (Carac®; Dermik Laboratories, Bridgewater, NJ)Antifolate inhibits proliferation; has shown anecdotal benefit in certain conditions characterized by hyperkeratotic lesions on the palms and soles


Summary

The addition of molecularly targeted agents to anticancer treatment has been found to cause both common and novel adverse reactions. HFSR is being increasingly recognized as a potential dose-limiting toxicity associated with sorafenib or sunitinib treatment that can result in discomfort, pain, decreased quality of life, and premature termination of a potentially effective cancer treatment. It is important to educate patients about potential dermatologic adverse effects associated with TKIs because limiting toxicity can help avoid treatment interruptions or dose reductions while improving ADLs.

The precise pathogenic mechanism of HFSR is currently not known, and there is no evidence-based protocol for treatment of HFSR. However, the increased clinical experience with these agents has resulted in a wealth of published articles describing empiric and symptomatic approaches that appear to help to prevent and manage HFSR. Frequent communication is necessary between the physician and patient, particularly 2–4 weeks from the initiation of therapy. Symptoms of HFSR should be recognized as early as possible. Providing the patient with a brochure about HFSR to refer to at home may facilitate the early identification of HFSR.

Patients should be advised of the “3C” approach to the management of TKI-associated HFSR: control calluses, comfort with cushions, and cover with creams. Creams should be applied after bathing and before going to bed; cotton gloves and socks should be worn to keep the cream on the hands and feet during the night.

Symptoms of HFSR typically are manageable with the implementation of supportive measures. If symptoms worsen, dose modification or interruption will result in a return to grade 0/1. Many patients can successfully be rechallenged with the full dose. Observations across multiple viewpoints have consistently shown that HFSR severity can be reduced in patients who are educated about HFSR and proactive about its detection and management.

Acknowledgments

All authors contributed equally to the development of this report. Editorial support was provided by Katherine Wright, PharmD, RPh, ISD, Wrighter Medical Education and Training, West Hills, CA; John A. Ibelli, CMPP, BelMed Professional Resources, New Rochelle, NY; and John D. Zoidis, MD, Bayer HealthCare Pharmaceuticals, Montville, NJ.

References1

1 C. Porta, C. Paglino, I. Imarisio and L. Bonomi, Uncovering Pandora's vase: the growing problem of new toxicities from novel anticancer agents: The case of sorafenib and sunitinib, Clin Exp Med 7 (2007), pp. 127–134. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

2 A.D. Lipworth, C. Robert and A.X. Zhu, Hand–foot syndrome (hand–foot skin reaction, palmar–plantar erythrodysesthesia): focus on sorafenib and sunitinib, Oncology 77 (2009), pp. 257–271 [19923864]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

3 M.E. Lacouture, S. Wu and C. Robert et al., Evolving strategies for the management of hand–foot skin reaction associated with the multitargeted kinase inhibitors sorafenib and sunitinib, Oncologist 13 (2008), pp. 1001–1011 [18779536]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (57)

4 S.M. Wilhelm, L. Adnane, P. Newell, A. Villanueva, J.M. Llovet and M. Lynch, Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling, Mol Cancer Ther 7 (2008), pp. 3129–3140 [18852116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)

5 D. Auclair, D. Miller and V. Yatsula et al., Antitumor activity of sorafenib in FLT3-driven leukemic cells, Leukemia 21 (2007), pp. 439–445 [17205056]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)

6 F. Carlomagno, S. Anaganti and T. Guida et al., BAY 43-9006 inhibition of oncogenic RET mutants, J Natl Cancer Inst 98 (2006), pp. 326–334 [16507829]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)

7 S.M. Wilhelm, C. Carter and L. Tang et al., BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis, Cancer Res 64 (2004), pp. 7099–7109 [15466206]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1142)

8 J.G. Christensen, A preclinical review of sunitinib, a multitargeted receptor tyrosine kinase inhibitor with anti-angiogenic and antitumour activities, Ann Oncol 10 (2007), pp. x3–x10 [17761721]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)

9 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib in advanced clear-cell renal-cell carcinoma, N Engl J Med 356 (2007), pp. 125–134 [17215530]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1425)

10 B. Escudier, T. Eisen and W.M. Stadler et al., Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III Treatment Approaches in Renal Cancer Global Evaluation Trial, J Clin Oncol 27 (2009), pp. 3312–3318 [19451442]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (123)

11 W.M. Stadler, R.A. Figlin and D.F. McDermott et al., Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in North America, Cancer 116 (2010), pp. 1272–1280 [20082451]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)

12 J. Beck, E. Bajetta and B. Escudier et al., A large open-label, non-comparative, phase III study of the multi-targeted kinase inhibitor sorafenib in European patients with advanced renal cell carcinoma, Eur J Cancer (suppl) (2007), p. 5 Abstract 4506. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)

13 J.M. Llovet, S. Ricci and V. Mazzaferro et al., Sorafenib in advanced hepatocellular carcinoma, N Engl J Med 359 (2008), pp. 378–390 [18650514]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (989)

14 A.L. Cheng, Y.K. Kang and Z. Chen et al., Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial, Lancet Oncol 10 (2009), pp. 25–34 [19095497]. Article |

PDF (195 K)
| View Record in Scopus | Cited By in Scopus (279)

15 G.D. Demetri, A.T. van Oosterom and C.R. Garrett et al., Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial, Lancet 368 (2006), pp. 1329–1338 [17046465]. Article |

PDF (200 K)
| View Record in Scopus | Cited By in Scopus (630)

16 G.D. Demetri, X. Huang and C.R. Garrett et al., Novel statistical analysis of long-term survival to account for crossover in a phase III trial of sunitinib (SU) vs placebo (PL) in advanced GIST after imatinib (IM) failure, J Clin Oncol (suppl) (2008), p. 26 Abstract 10524.

17 P. Reichardt, Y. Kang and W. Ruka et al., Detailed analysis of survival and safety with sunitinib (SU) in a worldwide treatment-use trial of patients with advanced GIST, J Clin Oncol (suppl) (2008), p. 26 Abstract 10548.

18 R.J. Motzer, T.E. Hutson and P. Tomczak et al., Sunitinib versus interferon alfa in metastatic renal-cell carcinoma, N Engl J Med 356 (2007), pp. 115–124 [17215529]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1520)

19R.J. Motzer, T.E. Hutson and P. Tomczak et al., Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 3584–3590 [19487381]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (194)

20 M.E. Gore, C. Szczylik and C. Porta et al., Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial, Lancet Oncol 10 (2009), pp. 757–763 [19615940]. Article |

PDF (142 K)
| View Record in Scopus | Cited By in Scopus (72)

21 G.R. Blumenschein Jr, U. Gatzemeier and F. Fossella et al., Phase II, multicenter, uncontrolled trial of single-agent sorafenib in patients with relapsed or refractory, advanced non-small-cell lung cancer, J Clin Oncol 27 (2009), pp. 4274–4280 [19652055]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)

22 H.J. Schütte, M. Schlemmer, C. Wendtner, P. Reichardt and G.D. Demetri, Sunitinib (SU) in a worldwide treatment-use trial of pts with advanced gastrointestinal stromal tumours (GIST): analysis of survival and safety data, Onkologie (suppl 4) (2008), p. 201 Abstract V582.

23 R.J. Motzer, B.I. Rini and R.M. Bukowski et al., Sunitinib in patients with metastatic renal cell carcinoma, JAMA 295 (2006), pp. 2516–2524 [16757724]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (586)

24 M.J. Ratain, T. Eisen and W.M. Stadler et al., Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma, J Clin Oncol 24 (2006), pp. 2505–2512 [16636341]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (544)

25 G.K. Abou-Alfa, L. Schwartz and S. Ricci et al., Phase II study of sorafenib in patients with advanced hepatocellular carcinoma, J Clin Oncol 24 (2006), pp. 4293–4300 [16908937]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (418)

26 T.E. Hutson, I.D. Davis and J.P. Machiels et al., Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma, J Clin Oncol 28 (2010), pp. 475–480 [20008644]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

27 C.N. Sternberg, I.D. Davis and J. Mardiak et al., Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial, J Clin Oncol 28 (2010), pp. 1061–1068 [20100962]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (100)

28 O. Rixe, R.M. Bukowski and M.D. Michaelson et al., Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study, Lancet Oncol 8 (2007), pp. 975–984 [17959415]. Article |

PDF (143 K)
| View Record in Scopus | Cited By in Scopus (132)

29 B.I. Rini, I. Tamaskar and P. Shaheen et al., Hypothyroidism in patients with metastatic renal cell carcinoma treated with sunitinib, J Natl Cancer Inst 99 (2007), pp. 81–83 [17202116]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (159)

30 B.I. Rini, G. Wilding and G. Hudes et al., Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma, J Clin Oncol 27 (2009), pp. 4462–4468 [19652060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (49)

31 H.S. Rugo, R.S. Herbst and G. Liu et al., Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: pharmacokinetic and clinical results, J Clin Oncol 23 (2005), pp. 5474–5483 [16027439]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (161)

32 National Cancer Institute, National Institutes of Health, US Department of Health and Human Services, Common Terminology Criteria for Adverse Events (CTCAE) Version 4.02. NIH publication 03-5410, National Institutes of Health, Bethesda, MD (2009).

33 L.S. Wood and B. Manchen, Sorafenib: a promising new targeted therapy for renal cell carcinoma, Clin J Oncol Nurs 11 (2007), pp. 649–656 [17962173]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)

34 C. Robert, C. Mateus, A. Spatz, J. Wechsler and B. Escudier, Dermatologic symptoms associated with the multikinase inhibitor sorafenib, J Am Acad Dermatol 60 (2009), pp. 299–305 [19028406]. Article |

PDF (296 K)
| View Record in Scopus | Cited By in Scopus (34)

35 J. Autier, B. Escudier, J. Wechsler, A. Spatz and C. Robert, Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor, Arch Dermatol 144 (2008), pp. 886–892 [18645140]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)

36 M.E. Lacouture, L.M. Reilly, P. Gerami and J. Guitart, Hand foot skin reaction in cancer patients treated with the multikinase inhibitors sorafenib and sunitinib, Ann Oncol 19 (2008), pp. 1955–1961 [18550575]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)

37 L.S. Wood, Managing the side effects of sorafenib and sunitinib, Community Oncol 3 (2006), pp. 558–562. View Record in Scopus | Cited By in Scopus (19)

38 C. Robert, J.C. Soria and A. Spatz et al., Cutaneous side-effects of kinase inhibitors and blocking antibodies, Lancet Oncol 6 (2005), pp. 491–500 [15992698]. Article |

PDF (533 K)
| View Record in Scopus | Cited By in Scopus (208)

39 M.W. Saif, I. Merikas, S. Tsimboukis and K. Syrigos, Erlotinib-induced skin rash: Pathogenesis, clinical significance and management in pancreatic cancer patients, JOP 9 (2008), pp. 267–274 [18469438]. View Record in Scopus | Cited By in Scopus (11)

40 R. Anderson, A. Jatoi, C. Robert, L.S. Wood, K.N. Keating and M.E. Lacouture, Search for evidence-based approaches for the prevention and palliation of hand–foot skin reaction (HFSR) caused by the multikinase inhibitors (MKIs), Oncologist 14 (2009), pp. 291–302 [19276294]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)

41 D.J. Gawkrodger, Dermatology: An Illustrated Colour Text, 4th ed, Churchill Livingstone Elsevier, Edinburgh (2008).

42 N.S. Azad, J.B. Aragon-Ching and W.L. Dahut et al., Hand–foot skin reaction increases with cumulative sorafenib dose and with combination anti-vascular endothelial growth factor therapy, Clin Cancer Res 15 (2009), pp. 1411–1416 [19228742]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)

43 N. Bhojani, C. Jeldres and J.J. Patard et al., Toxicities associated with the administration of sorafenib, sunitinib, and temsirolimus and their management in patients with metastatic renal cell carcinoma, Eur Urol 53 (2008), pp. 917–930 [18054825]. Article |

PDF (843 K)
| View Record in Scopus | Cited By in Scopus (81)

44 S. Sleijfer, I. Ray-Coquard and Z. Papai et al., Pazopanib, a multikinase angiogenesis inhibitor, in patients with relapsed or refractory advanced soft tissue sarcoma: a phase II study from the European Organisation for Research and Treatment of Cancer–Soft Tissue and Bone Sarcoma Group (EORTC study 62043), J Clin Oncol 27 (2009), pp. 3126–3132 [19451427]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)

45 L.S. Wood, H. Lemont and A. Jatoi et al., Practical considerations in the management of hand–foot skin reaction caused by multikinase inhibitors, Community Oncol 7 (2010), pp. 23–29. View Record in Scopus | Cited By in Scopus (3)

46 P. Esper, D. Gale and P. Muehlbauer, What kind of rash is it?: Deciphering the dermatologic toxicities of biologic and targeted therapies, Clin J Oncol Nurs 11 (2007), pp. 659–666 [17962174]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)

47 I. Hagemann and E. Proksch, Topical treatment by urea reduces epidermal hyperproliferation and induces differentiation in psoriasis, Acta Derm Venereol 76 (1996), pp. 353–356 [8891006]. View Record in Scopus | Cited By in Scopus (41)

48 M. Duvic, A.T. Asano, C. Hager and S. Mays, The pathogenesis of psoriasis and the mechanism of action of tazarotene, J Am Acad Dermatol 39 (1998), pp. S129–S133 [9777790]. Article |

PDF (205 K)
| View Record in Scopus | Cited By in Scopus (24)

49 W. Scheithauer, J. McKendrick and S. Begbie et al., Oral capecitabine as an alternative to i.v. 5-fluorouracil-based adjuvant therapy for colon cancer: safety results of a randomized, phase III trial, Ann Oncol 14 (2003), pp. 1735–1743 [14630678]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (172)

 

 

Conflicts of interest: For E. M., none to disclose. C. R. has participated in advisory boards for Bayer, Roche, Pfizer, Astrazeneca, and GSK in the field of melanoma treatment and the management of the cutaneous side effects of anticancer agents. C. P. has acted as a paid adviser or speaker for Bayer Schering Pharma, Pfizer Oncology, Hoffman La Roche, Novartis Pharma, GSK, and Wyeth Pharmaceuticals and has received research funding from Bayer Schering Pharma and Novartis Pharma.

Correspondence to: Elizabeth Manchen, RN, MS, OCN, Section of Hematology/Oncology, University of Chicago Medical Center, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637; telephone: (773) 702–4135


1 PubMed ID in brackets


Publications
Publications
Topics
Article Type
Display Headline
Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse
Display Headline
Management Of Tyrosine Kinase Inhibitor–Induced Hand–Foot Skin Reaction: Viewpoints from the Medical Oncologist, Dermatologist, and Oncology Nurse
Sections
Article Source

PURLs Copyright

Inside the Article