User login
Research and Reviews for the Practicing Oncologist
Tools for identifying, treating, monitoring, and preventing skin toxicities
The care of older cancer patients needs to be a priority for community oncologists
Undertreatment may contribute to deaths of older cancer patients
“I am wondering whether age is not a risk factor for [poor outcome in] lymphoma in general; it’s more a risk factor for not receiving proper treatment,” reflected Dr. Balducci, a professor of oncologic sciences at the University of South Florida College of Medicine and leader of the Senior Adult Oncology Program at the H. Lee Moffitt Cancer Center and Research Institute in Tampa.
Comorbidities must also be ascertained because of their potential impact on overall prognosis, treatment toxicity, drug interactions, and even cancer growth. Oncologists should look for features of so-called geriatric syndromes, such as spontaneous fractures, falls, or delirium precipitated by minor infections. “They generally are signs that indicate that the patient has not only low life expectancy, but also a poor tolerance of treatment,” he commented.
Oncologists should refer to the National Comprehensive Cancer Network (NCCN) clinical practice guidelines to help tailor chemotherapy in patients aged 65 years or older. Such recommendations include, for example, that the first dose be adjusted for renal function and that prophylactic filgrastim (Neupogen) or pegfilgrastim (Neulasta) be given to patients receiving moderately toxic regimens.
“I am wondering whether age is not a risk factor for [poor outcome in] lymphoma in general; it’s more a risk factor for not receiving proper treatment,” reflected Dr. Balducci, a professor of oncologic sciences at the University of South Florida College of Medicine and leader of the Senior Adult Oncology Program at the H. Lee Moffitt Cancer Center and Research Institute in Tampa.
Comorbidities must also be ascertained because of their potential impact on overall prognosis, treatment toxicity, drug interactions, and even cancer growth. Oncologists should look for features of so-called geriatric syndromes, such as spontaneous fractures, falls, or delirium precipitated by minor infections. “They generally are signs that indicate that the patient has not only low life expectancy, but also a poor tolerance of treatment,” he commented.
Oncologists should refer to the National Comprehensive Cancer Network (NCCN) clinical practice guidelines to help tailor chemotherapy in patients aged 65 years or older. Such recommendations include, for example, that the first dose be adjusted for renal function and that prophylactic filgrastim (Neupogen) or pegfilgrastim (Neulasta) be given to patients receiving moderately toxic regimens.
“I am wondering whether age is not a risk factor for [poor outcome in] lymphoma in general; it’s more a risk factor for not receiving proper treatment,” reflected Dr. Balducci, a professor of oncologic sciences at the University of South Florida College of Medicine and leader of the Senior Adult Oncology Program at the H. Lee Moffitt Cancer Center and Research Institute in Tampa.
Comorbidities must also be ascertained because of their potential impact on overall prognosis, treatment toxicity, drug interactions, and even cancer growth. Oncologists should look for features of so-called geriatric syndromes, such as spontaneous fractures, falls, or delirium precipitated by minor infections. “They generally are signs that indicate that the patient has not only low life expectancy, but also a poor tolerance of treatment,” he commented.
Oncologists should refer to the National Comprehensive Cancer Network (NCCN) clinical practice guidelines to help tailor chemotherapy in patients aged 65 years or older. Such recommendations include, for example, that the first dose be adjusted for renal function and that prophylactic filgrastim (Neupogen) or pegfilgrastim (Neulasta) be given to patients receiving moderately toxic regimens.
Optimal integration of EGFR inhibitors in advanced colorectal cancer
Cetuximab is a chimeric IgG1 (immunoglobulin G1) monoclonal antibody that binds to EGFR with high specificity and affinity.7 It was initially approved by the US Food and Drug Administration (FDA) in February 2004 based on response rate data in combination with irinotecan for irinotecan-refractory patients. Although originally used only in patients with EGFR-expressing tumors, a number of studies have since suggested that response is independent of EGFR staining, and this is no longer used as a selection criterion for EGFR-directed therapy.11–13 Rather than EGFR expression, it is KRAS mutation status that has emerged as the most important predictor for lack of benefit from anti-EGFR therapy. Multiple trials demonstrate either no benefit or inferior outcomes with the addition of EGFR-directed therapy in the approximately 40% of advanced colorectal cancer patients with activating KRAS mutations.
Panitumumab is a fully human IgG2 EGFR monoclonal antibody. Like cetuximab, it targets the extracellular domain of the EGFR and blocks downstream signaling, leading to antitumor effects.26 Because it is fully humanized, panitumumab does not have the same risk of hypersensitivity reactions seen with the chimeric antibody cetuximab. This may be especially important in some areas of the southeastern United States, where reactions to cetuximab occur at a higher frequency than has been reported in clinical trials.27 Panitumumab was approved for monotherapy of relapsed/refractory metastatic colorectal cancer by the FDA in September 2006. As with cetuximab, its use is now specified to tumors harboring wild-type KRAS.
ABOUT THE AUTHORS
Cetuximab is a chimeric IgG1 (immunoglobulin G1) monoclonal antibody that binds to EGFR with high specificity and affinity.7 It was initially approved by the US Food and Drug Administration (FDA) in February 2004 based on response rate data in combination with irinotecan for irinotecan-refractory patients. Although originally used only in patients with EGFR-expressing tumors, a number of studies have since suggested that response is independent of EGFR staining, and this is no longer used as a selection criterion for EGFR-directed therapy.11–13 Rather than EGFR expression, it is KRAS mutation status that has emerged as the most important predictor for lack of benefit from anti-EGFR therapy. Multiple trials demonstrate either no benefit or inferior outcomes with the addition of EGFR-directed therapy in the approximately 40% of advanced colorectal cancer patients with activating KRAS mutations.
Panitumumab is a fully human IgG2 EGFR monoclonal antibody. Like cetuximab, it targets the extracellular domain of the EGFR and blocks downstream signaling, leading to antitumor effects.26 Because it is fully humanized, panitumumab does not have the same risk of hypersensitivity reactions seen with the chimeric antibody cetuximab. This may be especially important in some areas of the southeastern United States, where reactions to cetuximab occur at a higher frequency than has been reported in clinical trials.27 Panitumumab was approved for monotherapy of relapsed/refractory metastatic colorectal cancer by the FDA in September 2006. As with cetuximab, its use is now specified to tumors harboring wild-type KRAS.
ABOUT THE AUTHORS
Cetuximab is a chimeric IgG1 (immunoglobulin G1) monoclonal antibody that binds to EGFR with high specificity and affinity.7 It was initially approved by the US Food and Drug Administration (FDA) in February 2004 based on response rate data in combination with irinotecan for irinotecan-refractory patients. Although originally used only in patients with EGFR-expressing tumors, a number of studies have since suggested that response is independent of EGFR staining, and this is no longer used as a selection criterion for EGFR-directed therapy.11–13 Rather than EGFR expression, it is KRAS mutation status that has emerged as the most important predictor for lack of benefit from anti-EGFR therapy. Multiple trials demonstrate either no benefit or inferior outcomes with the addition of EGFR-directed therapy in the approximately 40% of advanced colorectal cancer patients with activating KRAS mutations.
Panitumumab is a fully human IgG2 EGFR monoclonal antibody. Like cetuximab, it targets the extracellular domain of the EGFR and blocks downstream signaling, leading to antitumor effects.26 Because it is fully humanized, panitumumab does not have the same risk of hypersensitivity reactions seen with the chimeric antibody cetuximab. This may be especially important in some areas of the southeastern United States, where reactions to cetuximab occur at a higher frequency than has been reported in clinical trials.27 Panitumumab was approved for monotherapy of relapsed/refractory metastatic colorectal cancer by the FDA in September 2006. As with cetuximab, its use is now specified to tumors harboring wild-type KRAS.
ABOUT THE AUTHORS
Pomalidomide in lenalidomide-refractory multiple myeloma and carfilzomib in refractory and newly diagnosed multiple myeloma
--Jame Abraham, MD, Editor
1. Lacy MQ, Hayman SR, Gertz MA, et al. Pomalidomide (CC4047) plus low dose dexamethasone (Pom/dex) is active and well tolerated in lenalidomide refractory multiple myeloma (MM). Leukemia 2010;24:1934−1939.
2. Siegel DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label, single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
3. Jakubowiak AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma: initial results of phase I/II MMRC trial. Blood 2010;116:862.
4. Singhal SB, Siegel DSD, Martin T, et al. Pooled safety analysis from phase 1 and 2 studies of carfilzomib (CFZ) in patients with relapsed and/or refractory multiple myeloma (MM). Blood 2010;116:1954.
Noopur Raje, MD, Massachusetts General Hospital Cancer Center; Division of Hematology and Oncology, Massachusetts General Hospital; and Harvard Medical School, Boston, MA
Although multiple myeloma (MM) remains an incurable bone marrow
cancer, survival rates have improved markedly over the past decade. An
understanding of MM pathobiology (Figure 1) and improvement in stem cell
transplantation, better supportive care, and novel therapies with
higher efficacy and lower toxicity are all responsible for this
improvement. The availability of a rich pipeline of novel agents
undergoing early-phase clinical trials in MM is an exciting and active
area of research.1
Current treatment
Over the past several years, five therapeutic strategies have
received US Food and Drug Administration (FDA) approval either as
monotherapy or in combination for treating MM, with thalidomide
(Thalomid), lenalidomide (Revlimid), and bortezomib (Velcade) as
important backbone drugs in these approaches. In the upfront setting,
thalidomide with dexamethasone2 and bortezomib in combination with melphalan and prednisone3
increased the overall response rate and significantly prolonged time to
disease progression and are FDA approved. For treatment of relapsed MM,
bortezomib alone4 and in combination with pegylated liposomal doxorubicin (Doxil),5 as well as lenalidomide/dexamethasone,6
have been approved. Results of a recent phase III randomized clinical
trial suggest that lower doses of dexamethasone provide a survival
advantage, at least in the upfront setting, mainly due to the increased
toxicity of high doses of dexamethasone.7
The availability of these novel agents has not only provided us
with several treatment options but has had an important impact on the
overall survival of our patients. To improve upon current outcomes,
optimal combinations of bortezomib, thalidomide, and lenalidomide are
currently under evaluation in phase II/III clinical trials.
Novel approaches
The preceding review refers to recent data on pomalidomide, the
newest immunomodulatory drug (IMiD) analog, which has shown single-agent
activity in phase I studies and was subsequently tested in a phase II
trial in combination with low-dose dexamethasone in patients with
relapsed or refractory MM. Pomalidomide/dexamethasone was found to be
highly active and well tolerated, providing a clinical benefit in 47% of
patients and no grade 3 neuropathy. These findings have led to a large
phase II study, which has completed accrual and is awaiting analysis.
Another promising agent discussed here is the novel proteasome
inhibitor carfilzomib. Although bortezomib is an effective agent in MM,
about 20% of newly diagnosed patients are resistant to bortezomib, and,
ultimately, all patients relapse and develop resistance to the drug.
Carfilzomib irreversibly blocks chymotrypsin-like activity and in phase I
studies achieved more than 80% proteasome inhibition. Encouraging data
presented at the 2010 annual meeting of the American Society of
Hematology demonstrated that it was well tolerated and had an overall
clinical benefit rate of 36% in relapsed/refractory MM.8 In the upfront setting, carfilzomib combined with lenalidomide led to a 100% response rate.9
This combination with low-dose dexamethasone is currently
undergoing testing in a phase III registration trial. These data,
therefore, provide important therapeutic options among the armamentarium
of current and future antimyeloma therapies, helping transform MM into
an even more chronic disease than it is today and ultimately leading to a
cure.
References
1. Cirstea D, Vallet S, Raje N. Future novel single agent and combination therapies. Cancer J 2009;15:511-518.
2. Rajkumar
SV, Rosinol L, Hussein M, et al. Multicenter, randomized, double-blind,
placebo-controlled study of thalidomide plus dexamethasone compared
with dexamethasone as initial therapy for newly diagnosed multiple
myeloma. J Clin Oncol 2008;26:2171-2177.
3. San
Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and
prednisone for initial treatment of multiple myeloma. N Engl J Med
2008;359:906-917.
4. Richardson
PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose
dexamethasone for relapsed multiple myeloma. N Engl J Med
2005;352:2487-2498.
5. Orlowski
RZ, Nagler A, Sonneveld P, et al. Randomized phase III study of
pegylated liposomal doxorubicin plus bortezomib compared with bortezomib
alone in relapsed or refractory multiple myeloma: combination therapy
improves time to progression. J Clin Oncol 2007;25:3892-3901.
6. Dimopoulos
MA, Chen C, Spencer A, et al. Long-term follow-up on overall survival
from the MM-009 and MM-010 phase III trials of lenalidomide plus
dexamethasone in patients with relapsed or refractory multiple myeloma.
Leukemia 2009;23:2147-2152.
7. Rajkumar
SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose
dexamethasone versus lenalidomide plus low-dose dexamethasone as initial
therapy for newly diagnosed multiple myeloma: an open-label randomised
controlled trial. Lancet Oncol 2010;11:29-37.
8. Siegel
DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label,
single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with
relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
9. Jakubowiak
AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and
dexamethasone in newly diagnosed multiple myeloma: initial results of
phase I/II MMRC trial. Blood 2010;116:862.
Dr. Raje can be reached at [email protected].
Noopur Raje, MD, Massachusetts General Hospital Cancer Center; Division of Hematology and Oncology, Massachusetts General Hospital; and Harvard Medical School, Boston, MA
Although multiple myeloma (MM) remains an incurable bone marrow
cancer, survival rates have improved markedly over the past decade. An
understanding of MM pathobiology (Figure 1) and improvement in stem cell
transplantation, better supportive care, and novel therapies with
higher efficacy and lower toxicity are all responsible for this
improvement. The availability of a rich pipeline of novel agents
undergoing early-phase clinical trials in MM is an exciting and active
area of research.1
Current treatment
Over the past several years, five therapeutic strategies have
received US Food and Drug Administration (FDA) approval either as
monotherapy or in combination for treating MM, with thalidomide
(Thalomid), lenalidomide (Revlimid), and bortezomib (Velcade) as
important backbone drugs in these approaches. In the upfront setting,
thalidomide with dexamethasone2 and bortezomib in combination with melphalan and prednisone3
increased the overall response rate and significantly prolonged time to
disease progression and are FDA approved. For treatment of relapsed MM,
bortezomib alone4 and in combination with pegylated liposomal doxorubicin (Doxil),5 as well as lenalidomide/dexamethasone,6
have been approved. Results of a recent phase III randomized clinical
trial suggest that lower doses of dexamethasone provide a survival
advantage, at least in the upfront setting, mainly due to the increased
toxicity of high doses of dexamethasone.7
The availability of these novel agents has not only provided us
with several treatment options but has had an important impact on the
overall survival of our patients. To improve upon current outcomes,
optimal combinations of bortezomib, thalidomide, and lenalidomide are
currently under evaluation in phase II/III clinical trials.
Novel approaches
The preceding review refers to recent data on pomalidomide, the
newest immunomodulatory drug (IMiD) analog, which has shown single-agent
activity in phase I studies and was subsequently tested in a phase II
trial in combination with low-dose dexamethasone in patients with
relapsed or refractory MM. Pomalidomide/dexamethasone was found to be
highly active and well tolerated, providing a clinical benefit in 47% of
patients and no grade 3 neuropathy. These findings have led to a large
phase II study, which has completed accrual and is awaiting analysis.
Another promising agent discussed here is the novel proteasome
inhibitor carfilzomib. Although bortezomib is an effective agent in MM,
about 20% of newly diagnosed patients are resistant to bortezomib, and,
ultimately, all patients relapse and develop resistance to the drug.
Carfilzomib irreversibly blocks chymotrypsin-like activity and in phase I
studies achieved more than 80% proteasome inhibition. Encouraging data
presented at the 2010 annual meeting of the American Society of
Hematology demonstrated that it was well tolerated and had an overall
clinical benefit rate of 36% in relapsed/refractory MM.8 In the upfront setting, carfilzomib combined with lenalidomide led to a 100% response rate.9
This combination with low-dose dexamethasone is currently
undergoing testing in a phase III registration trial. These data,
therefore, provide important therapeutic options among the armamentarium
of current and future antimyeloma therapies, helping transform MM into
an even more chronic disease than it is today and ultimately leading to a
cure.
References
1. Cirstea D, Vallet S, Raje N. Future novel single agent and combination therapies. Cancer J 2009;15:511-518.
2. Rajkumar
SV, Rosinol L, Hussein M, et al. Multicenter, randomized, double-blind,
placebo-controlled study of thalidomide plus dexamethasone compared
with dexamethasone as initial therapy for newly diagnosed multiple
myeloma. J Clin Oncol 2008;26:2171-2177.
3. San
Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and
prednisone for initial treatment of multiple myeloma. N Engl J Med
2008;359:906-917.
4. Richardson
PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose
dexamethasone for relapsed multiple myeloma. N Engl J Med
2005;352:2487-2498.
5. Orlowski
RZ, Nagler A, Sonneveld P, et al. Randomized phase III study of
pegylated liposomal doxorubicin plus bortezomib compared with bortezomib
alone in relapsed or refractory multiple myeloma: combination therapy
improves time to progression. J Clin Oncol 2007;25:3892-3901.
6. Dimopoulos
MA, Chen C, Spencer A, et al. Long-term follow-up on overall survival
from the MM-009 and MM-010 phase III trials of lenalidomide plus
dexamethasone in patients with relapsed or refractory multiple myeloma.
Leukemia 2009;23:2147-2152.
7. Rajkumar
SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose
dexamethasone versus lenalidomide plus low-dose dexamethasone as initial
therapy for newly diagnosed multiple myeloma: an open-label randomised
controlled trial. Lancet Oncol 2010;11:29-37.
8. Siegel
DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label,
single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with
relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
9. Jakubowiak
AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and
dexamethasone in newly diagnosed multiple myeloma: initial results of
phase I/II MMRC trial. Blood 2010;116:862.
Dr. Raje can be reached at [email protected].
Noopur Raje, MD, Massachusetts General Hospital Cancer Center; Division of Hematology and Oncology, Massachusetts General Hospital; and Harvard Medical School, Boston, MA
Although multiple myeloma (MM) remains an incurable bone marrow
cancer, survival rates have improved markedly over the past decade. An
understanding of MM pathobiology (Figure 1) and improvement in stem cell
transplantation, better supportive care, and novel therapies with
higher efficacy and lower toxicity are all responsible for this
improvement. The availability of a rich pipeline of novel agents
undergoing early-phase clinical trials in MM is an exciting and active
area of research.1
Current treatment
Over the past several years, five therapeutic strategies have
received US Food and Drug Administration (FDA) approval either as
monotherapy or in combination for treating MM, with thalidomide
(Thalomid), lenalidomide (Revlimid), and bortezomib (Velcade) as
important backbone drugs in these approaches. In the upfront setting,
thalidomide with dexamethasone2 and bortezomib in combination with melphalan and prednisone3
increased the overall response rate and significantly prolonged time to
disease progression and are FDA approved. For treatment of relapsed MM,
bortezomib alone4 and in combination with pegylated liposomal doxorubicin (Doxil),5 as well as lenalidomide/dexamethasone,6
have been approved. Results of a recent phase III randomized clinical
trial suggest that lower doses of dexamethasone provide a survival
advantage, at least in the upfront setting, mainly due to the increased
toxicity of high doses of dexamethasone.7
The availability of these novel agents has not only provided us
with several treatment options but has had an important impact on the
overall survival of our patients. To improve upon current outcomes,
optimal combinations of bortezomib, thalidomide, and lenalidomide are
currently under evaluation in phase II/III clinical trials.
Novel approaches
The preceding review refers to recent data on pomalidomide, the
newest immunomodulatory drug (IMiD) analog, which has shown single-agent
activity in phase I studies and was subsequently tested in a phase II
trial in combination with low-dose dexamethasone in patients with
relapsed or refractory MM. Pomalidomide/dexamethasone was found to be
highly active and well tolerated, providing a clinical benefit in 47% of
patients and no grade 3 neuropathy. These findings have led to a large
phase II study, which has completed accrual and is awaiting analysis.
Another promising agent discussed here is the novel proteasome
inhibitor carfilzomib. Although bortezomib is an effective agent in MM,
about 20% of newly diagnosed patients are resistant to bortezomib, and,
ultimately, all patients relapse and develop resistance to the drug.
Carfilzomib irreversibly blocks chymotrypsin-like activity and in phase I
studies achieved more than 80% proteasome inhibition. Encouraging data
presented at the 2010 annual meeting of the American Society of
Hematology demonstrated that it was well tolerated and had an overall
clinical benefit rate of 36% in relapsed/refractory MM.8 In the upfront setting, carfilzomib combined with lenalidomide led to a 100% response rate.9
This combination with low-dose dexamethasone is currently
undergoing testing in a phase III registration trial. These data,
therefore, provide important therapeutic options among the armamentarium
of current and future antimyeloma therapies, helping transform MM into
an even more chronic disease than it is today and ultimately leading to a
cure.
References
1. Cirstea D, Vallet S, Raje N. Future novel single agent and combination therapies. Cancer J 2009;15:511-518.
2. Rajkumar
SV, Rosinol L, Hussein M, et al. Multicenter, randomized, double-blind,
placebo-controlled study of thalidomide plus dexamethasone compared
with dexamethasone as initial therapy for newly diagnosed multiple
myeloma. J Clin Oncol 2008;26:2171-2177.
3. San
Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and
prednisone for initial treatment of multiple myeloma. N Engl J Med
2008;359:906-917.
4. Richardson
PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose
dexamethasone for relapsed multiple myeloma. N Engl J Med
2005;352:2487-2498.
5. Orlowski
RZ, Nagler A, Sonneveld P, et al. Randomized phase III study of
pegylated liposomal doxorubicin plus bortezomib compared with bortezomib
alone in relapsed or refractory multiple myeloma: combination therapy
improves time to progression. J Clin Oncol 2007;25:3892-3901.
6. Dimopoulos
MA, Chen C, Spencer A, et al. Long-term follow-up on overall survival
from the MM-009 and MM-010 phase III trials of lenalidomide plus
dexamethasone in patients with relapsed or refractory multiple myeloma.
Leukemia 2009;23:2147-2152.
7. Rajkumar
SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose
dexamethasone versus lenalidomide plus low-dose dexamethasone as initial
therapy for newly diagnosed multiple myeloma: an open-label randomised
controlled trial. Lancet Oncol 2010;11:29-37.
8. Siegel
DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label,
single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with
relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
9. Jakubowiak
AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and
dexamethasone in newly diagnosed multiple myeloma: initial results of
phase I/II MMRC trial. Blood 2010;116:862.
Dr. Raje can be reached at [email protected].
--Jame Abraham, MD, Editor
1. Lacy MQ, Hayman SR, Gertz MA, et al. Pomalidomide (CC4047) plus low dose dexamethasone (Pom/dex) is active and well tolerated in lenalidomide refractory multiple myeloma (MM). Leukemia 2010;24:1934−1939.
2. Siegel DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label, single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
3. Jakubowiak AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma: initial results of phase I/II MMRC trial. Blood 2010;116:862.
4. Singhal SB, Siegel DSD, Martin T, et al. Pooled safety analysis from phase 1 and 2 studies of carfilzomib (CFZ) in patients with relapsed and/or refractory multiple myeloma (MM). Blood 2010;116:1954.
--Jame Abraham, MD, Editor
1. Lacy MQ, Hayman SR, Gertz MA, et al. Pomalidomide (CC4047) plus low dose dexamethasone (Pom/dex) is active and well tolerated in lenalidomide refractory multiple myeloma (MM). Leukemia 2010;24:1934−1939.
2. Siegel DSD, Martin T, Wang M, et al. Results of PX-171-003-A1, an open-label, single-arm, phase 2 study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM). Blood 2010;116:985.
3. Jakubowiak AJ, Dytfeld D, Jagannath S, et al. Carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma: initial results of phase I/II MMRC trial. Blood 2010;116:862.
4. Singhal SB, Siegel DSD, Martin T, et al. Pooled safety analysis from phase 1 and 2 studies of carfilzomib (CFZ) in patients with relapsed and/or refractory multiple myeloma (MM). Blood 2010;116:1954.
Treatment of recurrent high-grade gliomas
Myrna R. Rosenfeld, MD, PhD, Karen Albright, CRNP, and Amy A. Pruitt, MD
University of Pennsylvania School of Medicine, Philadelphia, PA
Manuscript received December 1, 2010; accepted April 15, 2011.
Correspondence to: Myrna R. Rosenfeld, MD, PhD, Department of Neurology, 3 W. Gates, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104; telephone: 215-746-2820; fax: 215-746-2818; e-mail: [email protected].
Despite treatment, glioblastoma (GB) inevitably recurs, and there is often no clear standard of care to follow. This article reviews the treatment options for recurrent GB and anaplastic gliomas. The three FDA-approved treatments for recurrent GB are biodegradable carmustine-impregnated wafers; bevacizumab; and the NovoTTF-100A System, which delivers low-intensity, alternating electrical fields to the tumor bed. Treatment decisions must take into consideration prior therapies, the extent and location of recurrence, and the patient’s general medical condition, as well as the rapidity of tumor growth, extent of edema, mass effect, need for steroids, and symptoms. New treatment strategies are emerging based on the identification of prognostic and predictive markers and defining distinct molecular subtypes of GB.
Glioblastoma (grade IV glioma, GB) is the most common of the primary malignant gliomas. Advances in the past decade have clarified that adjuvant treatment can improve quality of life and survival. The current standard of care for newly diagnosed patients with GB after optimal resection is focal radiation with concurrent (chemoradiation) and post-radiation temozolomide (Temodar). This regimen was shown to improve overall survival in a randomized phase III study.1 Furthermore, a subgroup of patients whose tumors were shown to have low levels of O6-methylguanine DNA methyltransferase (MGMT), the enzyme that repairs DNA damage due to temozolomide, showed overall survival of 46% at 2 years and 14% at 5 years.2,3
Biodegradable wafers impregnated with carmustine (Gliadel) implanted at the time of resection are another approved therapy for patients with newly diagnosed high-grade gliomas (GB and anaplastic gliomas, grade III glioma).4 Although short-term survivals for GB patients are similar with Gliadel compared to focal radiation with concurrent and post-radiation temozolomide, the latter results in superior long-term survival (1.9% at 56 months for Gliadel and 9.8% at 60 months for temozolomide).3,5 The nitrosoureas lomustine (CeeNU) and carmustine (BiCNU) were approved in the 1970s for use as single agents or in combination therapy in patients with glioma who had received surgery and/or radiation. These agents no longer have a clear role in the initial treatment of malignant glioma, although practitioners use them, often in combination therapy, for patients with recurrent GB and anaplastic oligodendrogliomas, a ¬chemosensitive subtype of glioma.6
Despite the use of the above approved therapies, GB invariably recurs. In the absence of effective treatment options, treatment approaches for recurrent GB have been varied. This article will review the most common accepted treatment options for recurrent GB and discuss emerging strategies. In the absence of a clear standard of care for newly diagnosed anaplastic gliomas, treatment at recurrence is largely dictated by prior therapies received, as noted below.
Tumor progression or treatment effect?
Glioblastoma recurrence is suspected when a previously stable patient develops recurrent or new neurologic signs and symptoms or when surveillance imaging (preferably MRI with gadolinium) shows increased tumor size or new enhancement likely associated with increased edema. However, clinical and imaging changes may result from perioperative complications such as infection or ischemia, a change in steroid use, or radiation necrosis (also called pseudoprogression). In fact, studies have shown that up to half of patients with presumed early tumor progression during or after chemoradiation actually have radiation necrosis (Figure 1).7 These data have led some clinicians to suggest that a minimum of 3 cycles of adjuvant temozolomide be given before a conclusion of tumor progression is made.
Several imaging modalities, such as magnetic resonance perfusion with or without spectroscopy and positron emission tomography, are used to help distinguish tumor recurrence from pseudoprogression but are not always reliable.8 In these cases, repeat imaging can be useful, although surgery may be necessary to relieve mass effect and obtain a tissue diagnosis.
Approved therapies for recurrent glioblastoma
There are currently three US Food and Drug Administration (FDA)–approved therapies for recurrent GB: Gliadel wafers; bevacizumab (Avastin), a humanized monoclonal antibody that sequesters vascular endothelial growth factor-A (VEGF); and the NovoTTF-100A System (NovoCure; Portsmouth, NH), which delivers low-intensity, alternating electrical fields (called tumor treatment fields) to the tumor bed that may inhibit cell growth by disrupting microtubule formation.9
The prospective randomized study that led to the approval of Gliadel demonstrated a modest increase in overall survival, from 23 weeks in those patients who received placebo wafers to 31 weeks for those receiving Gliadel.4 This study included recurrent anaplastic gliomas and GB, and the benefit in the GB subgroup was smaller than in the group as a whole. Furthermore, this and other studies predated the use of chemoradiation and adjuvant temozolomide, and it is unclear what benefit, if any, Gliadel offers in recurrent GB patients treated with prior temozolomide.
The approval of bevacizumab for recurrent GB in the United States was based on two clinical trials that evaluated bevacizumab as a single agent or combined with irinotecan in patients with tumor recurrence after initial treatment with chemoradiation and adjuvant temozolomide. The comparative study enrolled 167 patients, 85 in the bevacizumab-alone arm and 82 in the bevacizumab plus irinotecan arm.10 The objective response rate was 25.9% in patients who received bevacizumab monotherapy. There were no complete responses per outside review. The median duration of response was 4.2 months, and the 6-month progression-free survival (PFS-6) was 36.0 %. The single-arm study enrolled 56 patients with recurrent high-grade glioma.11 Objective response as determined by independent review was 19.6%. The median duration of response was 3.9 months.
The FDA approved bevacizumab for use as a single agent based on the improvement in objective response rate in these studies (albeit without improvement in disease-related symptoms or increased survival) and on the fact that patients receiving bevacizumab as a single agent had less toxicity than those receiving the combination with irinotecan and similar outcomes. Subsequent studies with bevacizumab as a single agent in recurrent GB have shown similar PFS-6 rates of between 29% and 42%; smaller studies in recurrent anaplastic gliomas reported response rates ranging between 34% and 68% and PFS-6 rates ranging between 32% and 68%.12,13
The NovoTTF-100A System received approval in the spring of 2011 based on the results of a single multinational study of 237 patients with recurrent GB.14 Patients were randomized to receive either the NovoTTF-100A System or chemotherapy of their physician’s choice. The NovoTTF device includes a battery pack (weighing about 6 pounds) and electrodes, which are placed on the scalp and designed to be worn for about 20 hours a day. The results showed that patients who used the NovoTTF-100A System had higher objective response rates (12% for NovoTTF compared with 6% in the chemotherapy group) and a favorable overall survival rate of 6.6 months compared with 6.0 months for the chemotherapy group. The device was well tolerated, with a significantly higher incidence of toxicities (hematologic and other) in the patients receiving chemotherapy.
Temozolomide is approved in Europe for recurrent high-grade gliomas, including both GB and anaplastic astrocytoma, but in the United States, it is only approved for recurrent anaplastic astrocytoma. These approvals were based on studies in mostly chemotherapy-naive patients, and although it is less clear that rechallenge with temozolomide is useful, as noted below, ongoing studies may clarify this issue.
Approach to the patient with recurrent GB
In the absence of enrollment in a clinical trial, which is encouraged for all glioma patients, the approach to the patient with recurrent GB should take into consideration prior therapies, extent of recurrence and location, and general medical condition. An initial consideration is whether the patient is a candidate for further resection and/or radiation. Re-resection can relieve mass effect and reduce the need for steroids, can provide histologic confirmation of the diagnosis, and likely can increase survival, although this has not been shown in randomized studies.15 For a patient with contraindications for further systemic chemotherapy and who is undergoing resection, Gliadel is a reasonable consideration, and it does not preclude use of bevacizumab or NovoTTF.
Re-irradiation with single-fraction or fractionated stereotactic radiation is feasible in patients with localized recurrent disease. Small, single-arm prospective studies and retrospective reviews suggest a benefit, and the focused delivery modalities reduce the dose to surrounding brain tissue, minimizing the risk of radiation toxicity.16 In some studies, low-dose temozolomide or bevacizumab was combined with re-irradiation with results suggesting both tolerability and efficacy.17 We have found re-irradiation to be a good alternative for those patients who develop profound and prolonged myelosuppression (usually thrombocytopenia) during initial treatment with temozolomide and who have tumor progression before recovery of the counts.
Bevacizumab is currently the most common single agent used for glioma recurrence and is usually dosed at 10 mg/kg every 2 weeks until disease progression, unacceptable toxicity, or the decision to discontinue care. Due to its potent anti-VEGF activity, which results in normalization of highly permeable tumor vessels, bevacizumab often produces rapid and marked reduction in edema and contrast enhancement on neuroimaging.18 This can produce rapid clinical and imaging responses, although whether this reflects true antiglioma activity remains under debate.
Resolution of mass effect and contrast enhancement can also coexist with progression of nonenhancing fluid-attenuated recovery (FLAIR) abnormality that reflects a phenotypically invasive tumor recurrence pattern, with GB co-option of normal cerebral vessels and diffuse, multilobar perivascular spread of tumor cells. The imaging changes can make evaluation of tumor response and progression difficult if one relies on the standard criteria of two-dimensional measurement of enhancing disease; new criteria that take into consideration nonenhancing signal abnormality changes have been proposed.19
Bevacizumab is well tolerated in the brain tumor population, with the same spectrum of side effects seen in other cancer populations (hemorrhage, thrombosis, hypertension, bowel perforation, impaired wound healing, and proteinuria).12,13 The incidence of life-threatening events, such as significant intracranial hemorrhage (3%) or thromboembolism (2%–12%), is within the expected range for the population and is not clearly increased by ¬bevacizumab.12
Several small series have reported that corticosteroid reductions were feasible in 33%–59% of patients with recurrent GB after bevacizumab treatment, and others have reported average corticosteroid dose reductions of 72% and 59%.11,13 This is an important benefit of bevacizumab, as chronic or high-dose corticosteroid use in patients with glioma is associated with significant morbidity. The ability of bevacizumab to control edema confounds the definition of tumor progression. Figure 2 shows a patient who had early neurologic improvement after initiation of bevacizumab and then remained clinically stable without corticosteroids over 14 months of therapy, despite slow continuous growth of the tumor mass.
Bevacizumab is also tolerated by older patients; there is an intriguing study suggesting that not only do older patients tolerate bevacizumab, they may also have increased benefit over younger patients (age separation: younger than 55 years or 55 years and older).20
Another option for some patients, prior to bevacizumab, is rechallenge with temozolomide at alternative dosing schedules, which result in prolonged exposure to higher cumulative doses than that achieved by standard 5-day dosing.21 Resistance to temozolomide occurs through direct repair of DNA by MGMT; a proposed mechanism to overcome resistance would be to deplete tumor-cell MGMT. Several studies have shown that prolonged exposure of peripheral blood mononuclear cells results in depletion of MGMT, and it has been suggested that this could also occur in glioma cells.22 Other studies suggest that prolonged exposure to temozolomide may be directly toxic to endothelial cells.23 These data provide a rationale for temozolomide rechallenge using alternative dose and dosing schedules that deliver higher culmulative doses over prolonged periods.
Commonly tried temozolomide schedules have been 21 days on/7 days off at doses of 75–100 mg/m2, 7 days on/7 days off at a dose of 150 mg/m2, and continuous daily dosing at 50 mg/m2 (Table 1). These schedules were well tolerated in these pretreated patients, with cumulative leukopenia after several cycles. Results have shown PFS-6 of 23%–48% with a suggestion (not supported by all studies) that best responses are seen in patients who were rechallenged after a treatment-free interval (from standard adjuvant temozolomide).24–27 Responses were also similar in patients with high and low levels of tumor MGMT, suggesting that these regimens may overcome -MGMT-mediated resistance.28
In addition to bevacizumab and temozolomide, lomustine, carmustine, irinotecan, cisplatin, and carboplatin have shown modest efficacy in studies as single agents or in combination regimens.29–31 The populations in these studies usually included both recurrent GB and anaplastic tumors, including oligodendrogliomas, and were carried out prior to standard use of chemoradiation and adjuvant temozolomide. Thus, it is difficult to extrapolate how these results would translate into today’s patient population. Interestingly, in a recent randomized phase III trial of recurrent GB (after prior temozolomide), lomustine was found to be superior to the investigational pan--VEFG receptor inhibitor cediranib.32
As noted, the NovoTTF-100A System was only recently approved, and experience is limited. Despite the need for patients to wear the device for 20 hours a day, and for intermittent adjustments to electrode placement at a clinic site, patient compliance in the study was good, and toxicities were minimal. It is clearly an option for patients with recurrent GB, but additional experience is needed to clarify the optimal time of use. The device is currently under study for the treatment of newly diagnosed GB; results of this study may help clarify the role of NovoTTF in treating this ¬malignancy.
In deciding which of the above strategies to use at first or even subsequent recurrences, we take into consideration the rapidity of tumor growth, extent of edema, mass effect, need for corticosteroids, and symptoms. Furthermore, to date there are no agents that improve outcomes when combined with bevacizumab or used after relapse on bevacizumab. This is often the last treatment regimen many patients receive before palliative end-of-life care. Thus, for a patient with a small, asymptomatic recurrence found on surveillance imaging and in the absence of an available clinical trial, we initiate therapy with any of the above-mentioned standard chemotherapeutic agents. At tumor progression and if chemotherapy is still tolerated, another agent may be tried. In contrast, the patient with a rapidly growing, large, or symptomatic recurrence requiring increasing doses of steroids will usually have an immediate clinical benefit from bevacizumab, which will improve quality of life.
Recurrent anaplastic gliomas
There is less consensus on how to treat anaplastic gliomas (anaplastic as¬tro¬cytoma, anaplastic oligodendroglioma, and mixed anaplastic oligoastrocytoma) at initial diagnosis and therefore even more variability in how these patients are treated at recurrence. The study that showed the benefit of chemoradiation and adjuvant temozolomide for newly diagnosed GB excluded patients with anaplastic tumors.1 A meta-analysis of clinical trials included adults with high-grade glioma who after initial surgery were treated with radiation plus chemotherapy (most often a nitrosourea) or radiation. The results only suggested that chemotherapy provided an additional survival benefit over radiotherapy alone for both GB and anaplastic patients.
Many cite the above data as the rationale for including temozolomide in the initial treatment of anaplastic tumors.33 Others, however, cite the results of NOA-04, a large study of patients with anaplastic glioma randomized to receive initial therapy with radiation or one of two chemotherapy regimens: procarbazine (Matulane), lomustine, and vincristine (PCV) or temozolomide.34 At tumor progression, patients who had received radiation were treated with either PCV or temozolomide, and those initially treated with either chemotherapy regimen were irradiated. This study demonstrated no difference in time to treatment failure or PFS among the three groups or any significant difference between the two chemotherapy regimens. Patients with anaplastic astrocytomas fared worse than those with anaplastic oligodendrogliomas or mixed tumors, suggesting that the latter groups may do well with initial chemotherapy only and then radiation at recurrence.
For patients with anaplastic tumors who have not previously been treated with temozolomide, studies do suggest that its use at recurrence is beneficial. In one phase II study, temozolomide-naive patients, or those who had previously received a nitrosourea, showed a 35% overall response rate and PFS-6 of 46% when treated with temozolomide at first recurrence.35 A recent study (RESCUE) evaluating a continuous low-dose temozolomide regimen of 50 mg/m2 in recurrent GB and anaplastic tumors demonstrated a PFS-6 of 35.7% for the anaplastic subgroup that contained patients who had previously had a variety of initial therapies, including chemoradiation.25
With the knowledge that the presence of chromosome 1p/19q codeletions in anaplastic tumors is prognostic for better outcomes,36 two ongoing studies will hopefully provide definitive answers for the treatment of recurrent anaplastic gliomas. The Chemoradiation and Adjuvant Temozolomide in Non-deleted Anaplastic Tumors (CATNON) study will randomly assign patients after surgery to receive either chemoradiation or radiation alone. Following this therapy, there is a second randomization to adjuvant temozolomide or observation only. The trial endpoint is overall survival. The phase III intergroup study of radiotherapy versus temozolomide alone versus radiotherapy with concomitant and adjuvant temozolomide for patients with 1p/19q codeleted anaplastic glioma (CODEL) will determine whether these patients with inherently better outcomes may do just as well with less aggressive therapy.
Several studies of bevacizumab have included recurrent anaplastic gliomas. Two studies of the combination of bevacizumab and irinotecan produced response rates of 55%–66% and PFS-6 of 56%–61%, suggesting activity.12,37 Extrapolating from the GB data, it is likely that single-agent bevacizumab would be efficacious and less toxic than this combination.
Emerging strategies
The identification of prognostic and predictive markers is paving the way for individualized treatment planning. In addition to the prognostic value of 1p/19q codeletions in anaplastic gliomas, the presence of MGMT promoter methylation in GB is likely predictive of response to temozolomide, although this is still under debate. There has been recent excitement about the demonstration that the presence of mutated isocitrate dehydrogenase 1 (IDH1) in gliomas is a robust independent factor associated with better outcome.38 For example, in a series of patients with anaplastic glioma, patients with the IDH1 mutation had a median survival four times longer than that of those without the mutation (81.1 months vs 19.4 months).39 This raises the question of the role of mutated IDH1 in glioma biology and makes it a potentially valuable therapeutic target.
Ongoing gene-expression profiling studies are showing that histologically indistinguishable GB can be clustered into distinct molecular subtypes, with widely different outcomes and responses to treatment.40 This likely contributed to the failure of past clinical trials, as the populations under study were, in fact, too diverse; potentially efficacious agents for one or more subtypes may have been overlooked. These studies are also identifying novel cellular targets such as MET, fibroblast growth factor receptor (FGFR), heat shock protein-90 (HSP-90), and hypoxia-inducible factor 1? (HIF1?).41 Other research is focusing on targets involved in glioma migration and invasion such as tenascin, the Src family of nonreceptor tyrosine kinases, the Rho family of small GTPases, and integrins. The role of glioma stem cells in glioma development and resistance to therapy is another emerging area of study and has led to the identification of specific glioma stem cell targets such as Notch and Sonic hedgehog.42
Conclusion
High-grade gliomas are challenging to treat, and there is often no clear standard of care. The Glioma Outcomes Project tracked clinical practice patterns and outcomes among North American patients with malignant glioma between 1997 and 2000.43 The results showed that patients treated at academic centers were significantly more likely to receive chemotherapy or radiation therapy, to participate in clinical trials, and to have longer survival times than those treated at community centers. Whether these results would be the same today, with the routine use of temozolomide and bevacizumab, is unclear, but they do support referral of these patients to centers with multispecialty clinics. This is, however, not always feasible, and patients may choose to stay close to home.
For patients with recurrent high-grade gliomas, there are several available therapeutic options, including operation, irradiation, and additional systemic therapies, which are available at most centers. Although the optimal sequence in which therapies should be given has not been clarified, these treatments can delay the onset of neurologic deficits and result in improved quality of life and likely prolonged survival. Additionally, the appropriate management of comorbidities such as seizures and brain edema is essential, and several pertinent reviews are available.44,45
References
- Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–996.
- Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997–1003.
- Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009;10:459–466.
- Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-Brain Tumor Treatment Group. Lancet 1995;345:1008–1012.
- Westphal M, Ram Z, Riddle V, et al. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien) 2006;148:269–275.
- Cairncross G, MacDonald D, Ludwin S, et al. Chemotherapy for anaplastic oligodendroglioma. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1994;12:2013–2021.
- Chamberlain MC, Glantz MJ, Chalmers L, et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol 2007;82:81–83.
- Clarke JL, Chang S. Pseudoprogression and pseudoresponse: challenges in brain tumor imaging. Curr Neurol Neurosci Rep 2009;9:241–246.
- Kirson ED, Dbaly V, Tovarys F, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A 2007;104:10152–10157.
- Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 2009;27:4733–4740.
- Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 2009;27:740–745.
- Vredenburgh JJ, Desjardins A, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 2007;25:4722–4729.
- Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology 2008;70:779–787.
- Stupp R, Kanner A, Engelhard H, et al. A prospective, randomized, open-label, phase III clinical trial of NovoTTF-100A versus best standard of care chemotherapy in patients with recurrent glioblastoma. J Clin Oncol 2010;28(18S):LBA2007.
- Park JK, Hodges T, Arko L, et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J Clin Oncol 2010;28:3838–3843.
- Combs SE, Thilmann C, Edler L, et al. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol 2005;23:8863–8869.
- Combs SE, Wagner J, Bischof M, et al. Radiochemotherapy in patients with primary glioblastoma comparing two temozolomide dose regimens. Int J Radiat Oncol Biol Phys 2008;71:999–1005.
- Pope WB, Lai A, Nghiemphu P, et al. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology 2006;66:1258–1260.
- Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:1963–1972.
- Nghiemphu PL, Liu W, Lee Y, et al. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology 2009;72:1217–1222.
- Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro Oncol 2009;11:69–79.
- Spiro TP, Liu L, Majka S, et al. Temozolomide: the effect of once- and twice-a-day dosing on tumor tissue levels of the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase. Clin Cancer Res 2001;7:2309–2317.
- Kurzen H, Schmitt S, Naher H, Mohler T. Inhibition of angiogenesis by non-toxic doses of temozolomide. Anticancer Drugs 2003;14:515–522.
- Wick A, Pascher C, Wick W, et al. Rechallenge with temozolomide in patients with recurrent gliomas. J Neurol 2009;256:734–741.
- Perry JR, Belanger K, Mason WP, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol 2010;28:2051–2057.
- Wick W, Steinbach JP, Kuker WM, Dichgans J, Bamberg M, Weller M. One week on/one week off: a novel active regimen of temozolomide for recurrent glioblastoma. Neurology 2004;62:2113–2115.
- Brandes AA, Tosoni A, Cavallo G, et al. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: phase II study from Gruppo Italiano Cooperativo di Neuro-oncologia (GICNO). Br J Cancer 2006;95:1155–1160.
- Wick A, Felsberg J, Steinbach JP, et al. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol 2007;25:3357–3361.
- Newton HB, Page MA, Junck L, et al. Intra-arterial cisplatin for the treatment of malignant gliomas. J Neurooncol 1989;7:39–45.
- Yung WK, Mechtler L, Gleason MJ. Intravenous carboplatin for recurrent malignant glioma: a phase II study. J Clin Oncol 1991;9:860–864.
- Gwak HS, Youn SM, Kwon AH, Lee SH, Kim JH, Rhee CH. ACNU-cisplatin continuous infusion chemotherapy as salvage therapy for recurrent glioblastomas: phase II study. J Neurooncol 2005;75:173–180.
- Batchelor T, Mulholland P, Neyns B, et al. The efficacy of cediranib as monotherapy and in combination with lomustine compared to lomustine alone in patients with recurrent glioblastoma: a phase III randomized study. J Neurooncol 2010;12(suppl 4):iv69–iv75.
- Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002;359:1011–1018.
- Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol 2009;27:5874–5880.
- Yung WK, Prados MD, Yaya-Tur R, et al. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group. J Clin Oncol 1999;17:2762–2771.
- Jenkins RB, Blair H, Ballman KV, et al. A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 2006;66:9852–9861.
- Desjardins A, Reardon DA, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent WHO grade 3 malignant gliomas. Clin Cancer Res 2008;14:7068–7073.
- Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–1812.
- Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol 2009;27:4150–4154.
- Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17:98–110.
- Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res 2009;29:5171–5184.
- Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY, Olson JJ. Exciting new advances in neuro-oncology: the avenue to a cure for malignant glioma. CA Cancer J Clin 2010;60:166–193.
- Chang SM, Parney IF, Huang W, et al. Patterns of care for adults with newly diagnosed malignant glioma. JAMA 2005;293:557–564.
- Pruitt AA. Treatment of medical complications in patients with brain tumors. Curr Treat Options Neurol 2005;7:323–336.
- Drappatz J, Schiff D, Kesari S, Norden AD, Wen PY. Medical management of brain tumor patients. Neurol Clin 2007;25:1035–1071, ix.
ABOUT THE AUTHORS
Affiliations: Dr. Rosenfeld is Adjunct Professor of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA; Ms. Albright is a Certified Registerd Nurse Practitioner at the University of Pennsylvania School of Medicine; and Dr. Pruitt is Associate Professor of Neurology at the University of Pennsylvania School of Medicine.
Conflicts of interest: The authors have nothing to disclose.
FIGURE 1 Pseudoprogression after chemoradiation. MRI of a biopsy-proven grade III astrocytoma before treatment (left panel). Three weeks after completion of chemoradiation, there is an increase in the size of the cystic component and enhancement (middle panel), which could be interpreted as tumor progression. After 4 cycles of post-radiation temozolomide, the right panel shows a decrease in size and enhancement, supporting the earlier imaging changes as reflective of pseudoprogression. All images are T1 post gadolinium with a 1.5 Tesla magnet.
FIGURE 2 Imaging changes with bevacizumab. This 57-year-old woman presented with a second recurrence of glioblastoma 2 years after chemoradiation and 6 cycles of adjuvant temozolomide and 6 months after 8 cycles of low-dose temozolomide. The images show post-contrast T1-weighted sequences (A, C, E) and fluid-attenuated recovery (FLAIR) sequences (B, D, F). Baseline images (A, B) show a left frontal mass with enhancement, edema, and some mass effect. Eight months later, after 4 cycles of bevacizumab (dosed every 2 weeks with 3 doses per cycle), there is decreased periventricular enhancement and edema (C) and improved FLAIR signal (D) but enlargement of the enhancing left frontal mass, which would meet standard definitions of tumor progression. The patient was neurologically stable and remained on treatment. Nine months later, after 5 additional cycles, she remained neurologically stable (and was not receiving corticosteroids). At this time, the mass has continued to increase in size (note: there is also invasion of the frontal sinus; E), with increasing FLAIR abnormality, likely reflecting a progressive infiltrating nonenhancing tumor (F).
Myrna R. Rosenfeld, MD, PhD, Karen Albright, CRNP, and Amy A. Pruitt, MD
University of Pennsylvania School of Medicine, Philadelphia, PA
Manuscript received December 1, 2010; accepted April 15, 2011.
Correspondence to: Myrna R. Rosenfeld, MD, PhD, Department of Neurology, 3 W. Gates, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104; telephone: 215-746-2820; fax: 215-746-2818; e-mail: [email protected].
Despite treatment, glioblastoma (GB) inevitably recurs, and there is often no clear standard of care to follow. This article reviews the treatment options for recurrent GB and anaplastic gliomas. The three FDA-approved treatments for recurrent GB are biodegradable carmustine-impregnated wafers; bevacizumab; and the NovoTTF-100A System, which delivers low-intensity, alternating electrical fields to the tumor bed. Treatment decisions must take into consideration prior therapies, the extent and location of recurrence, and the patient’s general medical condition, as well as the rapidity of tumor growth, extent of edema, mass effect, need for steroids, and symptoms. New treatment strategies are emerging based on the identification of prognostic and predictive markers and defining distinct molecular subtypes of GB.
Glioblastoma (grade IV glioma, GB) is the most common of the primary malignant gliomas. Advances in the past decade have clarified that adjuvant treatment can improve quality of life and survival. The current standard of care for newly diagnosed patients with GB after optimal resection is focal radiation with concurrent (chemoradiation) and post-radiation temozolomide (Temodar). This regimen was shown to improve overall survival in a randomized phase III study.1 Furthermore, a subgroup of patients whose tumors were shown to have low levels of O6-methylguanine DNA methyltransferase (MGMT), the enzyme that repairs DNA damage due to temozolomide, showed overall survival of 46% at 2 years and 14% at 5 years.2,3
Biodegradable wafers impregnated with carmustine (Gliadel) implanted at the time of resection are another approved therapy for patients with newly diagnosed high-grade gliomas (GB and anaplastic gliomas, grade III glioma).4 Although short-term survivals for GB patients are similar with Gliadel compared to focal radiation with concurrent and post-radiation temozolomide, the latter results in superior long-term survival (1.9% at 56 months for Gliadel and 9.8% at 60 months for temozolomide).3,5 The nitrosoureas lomustine (CeeNU) and carmustine (BiCNU) were approved in the 1970s for use as single agents or in combination therapy in patients with glioma who had received surgery and/or radiation. These agents no longer have a clear role in the initial treatment of malignant glioma, although practitioners use them, often in combination therapy, for patients with recurrent GB and anaplastic oligodendrogliomas, a ¬chemosensitive subtype of glioma.6
Despite the use of the above approved therapies, GB invariably recurs. In the absence of effective treatment options, treatment approaches for recurrent GB have been varied. This article will review the most common accepted treatment options for recurrent GB and discuss emerging strategies. In the absence of a clear standard of care for newly diagnosed anaplastic gliomas, treatment at recurrence is largely dictated by prior therapies received, as noted below.
Tumor progression or treatment effect?
Glioblastoma recurrence is suspected when a previously stable patient develops recurrent or new neurologic signs and symptoms or when surveillance imaging (preferably MRI with gadolinium) shows increased tumor size or new enhancement likely associated with increased edema. However, clinical and imaging changes may result from perioperative complications such as infection or ischemia, a change in steroid use, or radiation necrosis (also called pseudoprogression). In fact, studies have shown that up to half of patients with presumed early tumor progression during or after chemoradiation actually have radiation necrosis (Figure 1).7 These data have led some clinicians to suggest that a minimum of 3 cycles of adjuvant temozolomide be given before a conclusion of tumor progression is made.
Several imaging modalities, such as magnetic resonance perfusion with or without spectroscopy and positron emission tomography, are used to help distinguish tumor recurrence from pseudoprogression but are not always reliable.8 In these cases, repeat imaging can be useful, although surgery may be necessary to relieve mass effect and obtain a tissue diagnosis.
Approved therapies for recurrent glioblastoma
There are currently three US Food and Drug Administration (FDA)–approved therapies for recurrent GB: Gliadel wafers; bevacizumab (Avastin), a humanized monoclonal antibody that sequesters vascular endothelial growth factor-A (VEGF); and the NovoTTF-100A System (NovoCure; Portsmouth, NH), which delivers low-intensity, alternating electrical fields (called tumor treatment fields) to the tumor bed that may inhibit cell growth by disrupting microtubule formation.9
The prospective randomized study that led to the approval of Gliadel demonstrated a modest increase in overall survival, from 23 weeks in those patients who received placebo wafers to 31 weeks for those receiving Gliadel.4 This study included recurrent anaplastic gliomas and GB, and the benefit in the GB subgroup was smaller than in the group as a whole. Furthermore, this and other studies predated the use of chemoradiation and adjuvant temozolomide, and it is unclear what benefit, if any, Gliadel offers in recurrent GB patients treated with prior temozolomide.
The approval of bevacizumab for recurrent GB in the United States was based on two clinical trials that evaluated bevacizumab as a single agent or combined with irinotecan in patients with tumor recurrence after initial treatment with chemoradiation and adjuvant temozolomide. The comparative study enrolled 167 patients, 85 in the bevacizumab-alone arm and 82 in the bevacizumab plus irinotecan arm.10 The objective response rate was 25.9% in patients who received bevacizumab monotherapy. There were no complete responses per outside review. The median duration of response was 4.2 months, and the 6-month progression-free survival (PFS-6) was 36.0 %. The single-arm study enrolled 56 patients with recurrent high-grade glioma.11 Objective response as determined by independent review was 19.6%. The median duration of response was 3.9 months.
The FDA approved bevacizumab for use as a single agent based on the improvement in objective response rate in these studies (albeit without improvement in disease-related symptoms or increased survival) and on the fact that patients receiving bevacizumab as a single agent had less toxicity than those receiving the combination with irinotecan and similar outcomes. Subsequent studies with bevacizumab as a single agent in recurrent GB have shown similar PFS-6 rates of between 29% and 42%; smaller studies in recurrent anaplastic gliomas reported response rates ranging between 34% and 68% and PFS-6 rates ranging between 32% and 68%.12,13
The NovoTTF-100A System received approval in the spring of 2011 based on the results of a single multinational study of 237 patients with recurrent GB.14 Patients were randomized to receive either the NovoTTF-100A System or chemotherapy of their physician’s choice. The NovoTTF device includes a battery pack (weighing about 6 pounds) and electrodes, which are placed on the scalp and designed to be worn for about 20 hours a day. The results showed that patients who used the NovoTTF-100A System had higher objective response rates (12% for NovoTTF compared with 6% in the chemotherapy group) and a favorable overall survival rate of 6.6 months compared with 6.0 months for the chemotherapy group. The device was well tolerated, with a significantly higher incidence of toxicities (hematologic and other) in the patients receiving chemotherapy.
Temozolomide is approved in Europe for recurrent high-grade gliomas, including both GB and anaplastic astrocytoma, but in the United States, it is only approved for recurrent anaplastic astrocytoma. These approvals were based on studies in mostly chemotherapy-naive patients, and although it is less clear that rechallenge with temozolomide is useful, as noted below, ongoing studies may clarify this issue.
Approach to the patient with recurrent GB
In the absence of enrollment in a clinical trial, which is encouraged for all glioma patients, the approach to the patient with recurrent GB should take into consideration prior therapies, extent of recurrence and location, and general medical condition. An initial consideration is whether the patient is a candidate for further resection and/or radiation. Re-resection can relieve mass effect and reduce the need for steroids, can provide histologic confirmation of the diagnosis, and likely can increase survival, although this has not been shown in randomized studies.15 For a patient with contraindications for further systemic chemotherapy and who is undergoing resection, Gliadel is a reasonable consideration, and it does not preclude use of bevacizumab or NovoTTF.
Re-irradiation with single-fraction or fractionated stereotactic radiation is feasible in patients with localized recurrent disease. Small, single-arm prospective studies and retrospective reviews suggest a benefit, and the focused delivery modalities reduce the dose to surrounding brain tissue, minimizing the risk of radiation toxicity.16 In some studies, low-dose temozolomide or bevacizumab was combined with re-irradiation with results suggesting both tolerability and efficacy.17 We have found re-irradiation to be a good alternative for those patients who develop profound and prolonged myelosuppression (usually thrombocytopenia) during initial treatment with temozolomide and who have tumor progression before recovery of the counts.
Bevacizumab is currently the most common single agent used for glioma recurrence and is usually dosed at 10 mg/kg every 2 weeks until disease progression, unacceptable toxicity, or the decision to discontinue care. Due to its potent anti-VEGF activity, which results in normalization of highly permeable tumor vessels, bevacizumab often produces rapid and marked reduction in edema and contrast enhancement on neuroimaging.18 This can produce rapid clinical and imaging responses, although whether this reflects true antiglioma activity remains under debate.
Resolution of mass effect and contrast enhancement can also coexist with progression of nonenhancing fluid-attenuated recovery (FLAIR) abnormality that reflects a phenotypically invasive tumor recurrence pattern, with GB co-option of normal cerebral vessels and diffuse, multilobar perivascular spread of tumor cells. The imaging changes can make evaluation of tumor response and progression difficult if one relies on the standard criteria of two-dimensional measurement of enhancing disease; new criteria that take into consideration nonenhancing signal abnormality changes have been proposed.19
Bevacizumab is well tolerated in the brain tumor population, with the same spectrum of side effects seen in other cancer populations (hemorrhage, thrombosis, hypertension, bowel perforation, impaired wound healing, and proteinuria).12,13 The incidence of life-threatening events, such as significant intracranial hemorrhage (3%) or thromboembolism (2%–12%), is within the expected range for the population and is not clearly increased by ¬bevacizumab.12
Several small series have reported that corticosteroid reductions were feasible in 33%–59% of patients with recurrent GB after bevacizumab treatment, and others have reported average corticosteroid dose reductions of 72% and 59%.11,13 This is an important benefit of bevacizumab, as chronic or high-dose corticosteroid use in patients with glioma is associated with significant morbidity. The ability of bevacizumab to control edema confounds the definition of tumor progression. Figure 2 shows a patient who had early neurologic improvement after initiation of bevacizumab and then remained clinically stable without corticosteroids over 14 months of therapy, despite slow continuous growth of the tumor mass.
Bevacizumab is also tolerated by older patients; there is an intriguing study suggesting that not only do older patients tolerate bevacizumab, they may also have increased benefit over younger patients (age separation: younger than 55 years or 55 years and older).20
Another option for some patients, prior to bevacizumab, is rechallenge with temozolomide at alternative dosing schedules, which result in prolonged exposure to higher cumulative doses than that achieved by standard 5-day dosing.21 Resistance to temozolomide occurs through direct repair of DNA by MGMT; a proposed mechanism to overcome resistance would be to deplete tumor-cell MGMT. Several studies have shown that prolonged exposure of peripheral blood mononuclear cells results in depletion of MGMT, and it has been suggested that this could also occur in glioma cells.22 Other studies suggest that prolonged exposure to temozolomide may be directly toxic to endothelial cells.23 These data provide a rationale for temozolomide rechallenge using alternative dose and dosing schedules that deliver higher culmulative doses over prolonged periods.
Commonly tried temozolomide schedules have been 21 days on/7 days off at doses of 75–100 mg/m2, 7 days on/7 days off at a dose of 150 mg/m2, and continuous daily dosing at 50 mg/m2 (Table 1). These schedules were well tolerated in these pretreated patients, with cumulative leukopenia after several cycles. Results have shown PFS-6 of 23%–48% with a suggestion (not supported by all studies) that best responses are seen in patients who were rechallenged after a treatment-free interval (from standard adjuvant temozolomide).24–27 Responses were also similar in patients with high and low levels of tumor MGMT, suggesting that these regimens may overcome -MGMT-mediated resistance.28
In addition to bevacizumab and temozolomide, lomustine, carmustine, irinotecan, cisplatin, and carboplatin have shown modest efficacy in studies as single agents or in combination regimens.29–31 The populations in these studies usually included both recurrent GB and anaplastic tumors, including oligodendrogliomas, and were carried out prior to standard use of chemoradiation and adjuvant temozolomide. Thus, it is difficult to extrapolate how these results would translate into today’s patient population. Interestingly, in a recent randomized phase III trial of recurrent GB (after prior temozolomide), lomustine was found to be superior to the investigational pan--VEFG receptor inhibitor cediranib.32
As noted, the NovoTTF-100A System was only recently approved, and experience is limited. Despite the need for patients to wear the device for 20 hours a day, and for intermittent adjustments to electrode placement at a clinic site, patient compliance in the study was good, and toxicities were minimal. It is clearly an option for patients with recurrent GB, but additional experience is needed to clarify the optimal time of use. The device is currently under study for the treatment of newly diagnosed GB; results of this study may help clarify the role of NovoTTF in treating this ¬malignancy.
In deciding which of the above strategies to use at first or even subsequent recurrences, we take into consideration the rapidity of tumor growth, extent of edema, mass effect, need for corticosteroids, and symptoms. Furthermore, to date there are no agents that improve outcomes when combined with bevacizumab or used after relapse on bevacizumab. This is often the last treatment regimen many patients receive before palliative end-of-life care. Thus, for a patient with a small, asymptomatic recurrence found on surveillance imaging and in the absence of an available clinical trial, we initiate therapy with any of the above-mentioned standard chemotherapeutic agents. At tumor progression and if chemotherapy is still tolerated, another agent may be tried. In contrast, the patient with a rapidly growing, large, or symptomatic recurrence requiring increasing doses of steroids will usually have an immediate clinical benefit from bevacizumab, which will improve quality of life.
Recurrent anaplastic gliomas
There is less consensus on how to treat anaplastic gliomas (anaplastic as¬tro¬cytoma, anaplastic oligodendroglioma, and mixed anaplastic oligoastrocytoma) at initial diagnosis and therefore even more variability in how these patients are treated at recurrence. The study that showed the benefit of chemoradiation and adjuvant temozolomide for newly diagnosed GB excluded patients with anaplastic tumors.1 A meta-analysis of clinical trials included adults with high-grade glioma who after initial surgery were treated with radiation plus chemotherapy (most often a nitrosourea) or radiation. The results only suggested that chemotherapy provided an additional survival benefit over radiotherapy alone for both GB and anaplastic patients.
Many cite the above data as the rationale for including temozolomide in the initial treatment of anaplastic tumors.33 Others, however, cite the results of NOA-04, a large study of patients with anaplastic glioma randomized to receive initial therapy with radiation or one of two chemotherapy regimens: procarbazine (Matulane), lomustine, and vincristine (PCV) or temozolomide.34 At tumor progression, patients who had received radiation were treated with either PCV or temozolomide, and those initially treated with either chemotherapy regimen were irradiated. This study demonstrated no difference in time to treatment failure or PFS among the three groups or any significant difference between the two chemotherapy regimens. Patients with anaplastic astrocytomas fared worse than those with anaplastic oligodendrogliomas or mixed tumors, suggesting that the latter groups may do well with initial chemotherapy only and then radiation at recurrence.
For patients with anaplastic tumors who have not previously been treated with temozolomide, studies do suggest that its use at recurrence is beneficial. In one phase II study, temozolomide-naive patients, or those who had previously received a nitrosourea, showed a 35% overall response rate and PFS-6 of 46% when treated with temozolomide at first recurrence.35 A recent study (RESCUE) evaluating a continuous low-dose temozolomide regimen of 50 mg/m2 in recurrent GB and anaplastic tumors demonstrated a PFS-6 of 35.7% for the anaplastic subgroup that contained patients who had previously had a variety of initial therapies, including chemoradiation.25
With the knowledge that the presence of chromosome 1p/19q codeletions in anaplastic tumors is prognostic for better outcomes,36 two ongoing studies will hopefully provide definitive answers for the treatment of recurrent anaplastic gliomas. The Chemoradiation and Adjuvant Temozolomide in Non-deleted Anaplastic Tumors (CATNON) study will randomly assign patients after surgery to receive either chemoradiation or radiation alone. Following this therapy, there is a second randomization to adjuvant temozolomide or observation only. The trial endpoint is overall survival. The phase III intergroup study of radiotherapy versus temozolomide alone versus radiotherapy with concomitant and adjuvant temozolomide for patients with 1p/19q codeleted anaplastic glioma (CODEL) will determine whether these patients with inherently better outcomes may do just as well with less aggressive therapy.
Several studies of bevacizumab have included recurrent anaplastic gliomas. Two studies of the combination of bevacizumab and irinotecan produced response rates of 55%–66% and PFS-6 of 56%–61%, suggesting activity.12,37 Extrapolating from the GB data, it is likely that single-agent bevacizumab would be efficacious and less toxic than this combination.
Emerging strategies
The identification of prognostic and predictive markers is paving the way for individualized treatment planning. In addition to the prognostic value of 1p/19q codeletions in anaplastic gliomas, the presence of MGMT promoter methylation in GB is likely predictive of response to temozolomide, although this is still under debate. There has been recent excitement about the demonstration that the presence of mutated isocitrate dehydrogenase 1 (IDH1) in gliomas is a robust independent factor associated with better outcome.38 For example, in a series of patients with anaplastic glioma, patients with the IDH1 mutation had a median survival four times longer than that of those without the mutation (81.1 months vs 19.4 months).39 This raises the question of the role of mutated IDH1 in glioma biology and makes it a potentially valuable therapeutic target.
Ongoing gene-expression profiling studies are showing that histologically indistinguishable GB can be clustered into distinct molecular subtypes, with widely different outcomes and responses to treatment.40 This likely contributed to the failure of past clinical trials, as the populations under study were, in fact, too diverse; potentially efficacious agents for one or more subtypes may have been overlooked. These studies are also identifying novel cellular targets such as MET, fibroblast growth factor receptor (FGFR), heat shock protein-90 (HSP-90), and hypoxia-inducible factor 1? (HIF1?).41 Other research is focusing on targets involved in glioma migration and invasion such as tenascin, the Src family of nonreceptor tyrosine kinases, the Rho family of small GTPases, and integrins. The role of glioma stem cells in glioma development and resistance to therapy is another emerging area of study and has led to the identification of specific glioma stem cell targets such as Notch and Sonic hedgehog.42
Conclusion
High-grade gliomas are challenging to treat, and there is often no clear standard of care. The Glioma Outcomes Project tracked clinical practice patterns and outcomes among North American patients with malignant glioma between 1997 and 2000.43 The results showed that patients treated at academic centers were significantly more likely to receive chemotherapy or radiation therapy, to participate in clinical trials, and to have longer survival times than those treated at community centers. Whether these results would be the same today, with the routine use of temozolomide and bevacizumab, is unclear, but they do support referral of these patients to centers with multispecialty clinics. This is, however, not always feasible, and patients may choose to stay close to home.
For patients with recurrent high-grade gliomas, there are several available therapeutic options, including operation, irradiation, and additional systemic therapies, which are available at most centers. Although the optimal sequence in which therapies should be given has not been clarified, these treatments can delay the onset of neurologic deficits and result in improved quality of life and likely prolonged survival. Additionally, the appropriate management of comorbidities such as seizures and brain edema is essential, and several pertinent reviews are available.44,45
References
- Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–996.
- Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997–1003.
- Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009;10:459–466.
- Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-Brain Tumor Treatment Group. Lancet 1995;345:1008–1012.
- Westphal M, Ram Z, Riddle V, et al. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien) 2006;148:269–275.
- Cairncross G, MacDonald D, Ludwin S, et al. Chemotherapy for anaplastic oligodendroglioma. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1994;12:2013–2021.
- Chamberlain MC, Glantz MJ, Chalmers L, et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol 2007;82:81–83.
- Clarke JL, Chang S. Pseudoprogression and pseudoresponse: challenges in brain tumor imaging. Curr Neurol Neurosci Rep 2009;9:241–246.
- Kirson ED, Dbaly V, Tovarys F, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A 2007;104:10152–10157.
- Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 2009;27:4733–4740.
- Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 2009;27:740–745.
- Vredenburgh JJ, Desjardins A, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 2007;25:4722–4729.
- Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology 2008;70:779–787.
- Stupp R, Kanner A, Engelhard H, et al. A prospective, randomized, open-label, phase III clinical trial of NovoTTF-100A versus best standard of care chemotherapy in patients with recurrent glioblastoma. J Clin Oncol 2010;28(18S):LBA2007.
- Park JK, Hodges T, Arko L, et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J Clin Oncol 2010;28:3838–3843.
- Combs SE, Thilmann C, Edler L, et al. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol 2005;23:8863–8869.
- Combs SE, Wagner J, Bischof M, et al. Radiochemotherapy in patients with primary glioblastoma comparing two temozolomide dose regimens. Int J Radiat Oncol Biol Phys 2008;71:999–1005.
- Pope WB, Lai A, Nghiemphu P, et al. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology 2006;66:1258–1260.
- Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:1963–1972.
- Nghiemphu PL, Liu W, Lee Y, et al. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology 2009;72:1217–1222.
- Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro Oncol 2009;11:69–79.
- Spiro TP, Liu L, Majka S, et al. Temozolomide: the effect of once- and twice-a-day dosing on tumor tissue levels of the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase. Clin Cancer Res 2001;7:2309–2317.
- Kurzen H, Schmitt S, Naher H, Mohler T. Inhibition of angiogenesis by non-toxic doses of temozolomide. Anticancer Drugs 2003;14:515–522.
- Wick A, Pascher C, Wick W, et al. Rechallenge with temozolomide in patients with recurrent gliomas. J Neurol 2009;256:734–741.
- Perry JR, Belanger K, Mason WP, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol 2010;28:2051–2057.
- Wick W, Steinbach JP, Kuker WM, Dichgans J, Bamberg M, Weller M. One week on/one week off: a novel active regimen of temozolomide for recurrent glioblastoma. Neurology 2004;62:2113–2115.
- Brandes AA, Tosoni A, Cavallo G, et al. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: phase II study from Gruppo Italiano Cooperativo di Neuro-oncologia (GICNO). Br J Cancer 2006;95:1155–1160.
- Wick A, Felsberg J, Steinbach JP, et al. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol 2007;25:3357–3361.
- Newton HB, Page MA, Junck L, et al. Intra-arterial cisplatin for the treatment of malignant gliomas. J Neurooncol 1989;7:39–45.
- Yung WK, Mechtler L, Gleason MJ. Intravenous carboplatin for recurrent malignant glioma: a phase II study. J Clin Oncol 1991;9:860–864.
- Gwak HS, Youn SM, Kwon AH, Lee SH, Kim JH, Rhee CH. ACNU-cisplatin continuous infusion chemotherapy as salvage therapy for recurrent glioblastomas: phase II study. J Neurooncol 2005;75:173–180.
- Batchelor T, Mulholland P, Neyns B, et al. The efficacy of cediranib as monotherapy and in combination with lomustine compared to lomustine alone in patients with recurrent glioblastoma: a phase III randomized study. J Neurooncol 2010;12(suppl 4):iv69–iv75.
- Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002;359:1011–1018.
- Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol 2009;27:5874–5880.
- Yung WK, Prados MD, Yaya-Tur R, et al. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group. J Clin Oncol 1999;17:2762–2771.
- Jenkins RB, Blair H, Ballman KV, et al. A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 2006;66:9852–9861.
- Desjardins A, Reardon DA, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent WHO grade 3 malignant gliomas. Clin Cancer Res 2008;14:7068–7073.
- Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–1812.
- Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol 2009;27:4150–4154.
- Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17:98–110.
- Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res 2009;29:5171–5184.
- Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY, Olson JJ. Exciting new advances in neuro-oncology: the avenue to a cure for malignant glioma. CA Cancer J Clin 2010;60:166–193.
- Chang SM, Parney IF, Huang W, et al. Patterns of care for adults with newly diagnosed malignant glioma. JAMA 2005;293:557–564.
- Pruitt AA. Treatment of medical complications in patients with brain tumors. Curr Treat Options Neurol 2005;7:323–336.
- Drappatz J, Schiff D, Kesari S, Norden AD, Wen PY. Medical management of brain tumor patients. Neurol Clin 2007;25:1035–1071, ix.
ABOUT THE AUTHORS
Affiliations: Dr. Rosenfeld is Adjunct Professor of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA; Ms. Albright is a Certified Registerd Nurse Practitioner at the University of Pennsylvania School of Medicine; and Dr. Pruitt is Associate Professor of Neurology at the University of Pennsylvania School of Medicine.
Conflicts of interest: The authors have nothing to disclose.
FIGURE 1 Pseudoprogression after chemoradiation. MRI of a biopsy-proven grade III astrocytoma before treatment (left panel). Three weeks after completion of chemoradiation, there is an increase in the size of the cystic component and enhancement (middle panel), which could be interpreted as tumor progression. After 4 cycles of post-radiation temozolomide, the right panel shows a decrease in size and enhancement, supporting the earlier imaging changes as reflective of pseudoprogression. All images are T1 post gadolinium with a 1.5 Tesla magnet.
FIGURE 2 Imaging changes with bevacizumab. This 57-year-old woman presented with a second recurrence of glioblastoma 2 years after chemoradiation and 6 cycles of adjuvant temozolomide and 6 months after 8 cycles of low-dose temozolomide. The images show post-contrast T1-weighted sequences (A, C, E) and fluid-attenuated recovery (FLAIR) sequences (B, D, F). Baseline images (A, B) show a left frontal mass with enhancement, edema, and some mass effect. Eight months later, after 4 cycles of bevacizumab (dosed every 2 weeks with 3 doses per cycle), there is decreased periventricular enhancement and edema (C) and improved FLAIR signal (D) but enlargement of the enhancing left frontal mass, which would meet standard definitions of tumor progression. The patient was neurologically stable and remained on treatment. Nine months later, after 5 additional cycles, she remained neurologically stable (and was not receiving corticosteroids). At this time, the mass has continued to increase in size (note: there is also invasion of the frontal sinus; E), with increasing FLAIR abnormality, likely reflecting a progressive infiltrating nonenhancing tumor (F).
Myrna R. Rosenfeld, MD, PhD, Karen Albright, CRNP, and Amy A. Pruitt, MD
University of Pennsylvania School of Medicine, Philadelphia, PA
Manuscript received December 1, 2010; accepted April 15, 2011.
Correspondence to: Myrna R. Rosenfeld, MD, PhD, Department of Neurology, 3 W. Gates, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104; telephone: 215-746-2820; fax: 215-746-2818; e-mail: [email protected].
Despite treatment, glioblastoma (GB) inevitably recurs, and there is often no clear standard of care to follow. This article reviews the treatment options for recurrent GB and anaplastic gliomas. The three FDA-approved treatments for recurrent GB are biodegradable carmustine-impregnated wafers; bevacizumab; and the NovoTTF-100A System, which delivers low-intensity, alternating electrical fields to the tumor bed. Treatment decisions must take into consideration prior therapies, the extent and location of recurrence, and the patient’s general medical condition, as well as the rapidity of tumor growth, extent of edema, mass effect, need for steroids, and symptoms. New treatment strategies are emerging based on the identification of prognostic and predictive markers and defining distinct molecular subtypes of GB.
Glioblastoma (grade IV glioma, GB) is the most common of the primary malignant gliomas. Advances in the past decade have clarified that adjuvant treatment can improve quality of life and survival. The current standard of care for newly diagnosed patients with GB after optimal resection is focal radiation with concurrent (chemoradiation) and post-radiation temozolomide (Temodar). This regimen was shown to improve overall survival in a randomized phase III study.1 Furthermore, a subgroup of patients whose tumors were shown to have low levels of O6-methylguanine DNA methyltransferase (MGMT), the enzyme that repairs DNA damage due to temozolomide, showed overall survival of 46% at 2 years and 14% at 5 years.2,3
Biodegradable wafers impregnated with carmustine (Gliadel) implanted at the time of resection are another approved therapy for patients with newly diagnosed high-grade gliomas (GB and anaplastic gliomas, grade III glioma).4 Although short-term survivals for GB patients are similar with Gliadel compared to focal radiation with concurrent and post-radiation temozolomide, the latter results in superior long-term survival (1.9% at 56 months for Gliadel and 9.8% at 60 months for temozolomide).3,5 The nitrosoureas lomustine (CeeNU) and carmustine (BiCNU) were approved in the 1970s for use as single agents or in combination therapy in patients with glioma who had received surgery and/or radiation. These agents no longer have a clear role in the initial treatment of malignant glioma, although practitioners use them, often in combination therapy, for patients with recurrent GB and anaplastic oligodendrogliomas, a ¬chemosensitive subtype of glioma.6
Despite the use of the above approved therapies, GB invariably recurs. In the absence of effective treatment options, treatment approaches for recurrent GB have been varied. This article will review the most common accepted treatment options for recurrent GB and discuss emerging strategies. In the absence of a clear standard of care for newly diagnosed anaplastic gliomas, treatment at recurrence is largely dictated by prior therapies received, as noted below.
Tumor progression or treatment effect?
Glioblastoma recurrence is suspected when a previously stable patient develops recurrent or new neurologic signs and symptoms or when surveillance imaging (preferably MRI with gadolinium) shows increased tumor size or new enhancement likely associated with increased edema. However, clinical and imaging changes may result from perioperative complications such as infection or ischemia, a change in steroid use, or radiation necrosis (also called pseudoprogression). In fact, studies have shown that up to half of patients with presumed early tumor progression during or after chemoradiation actually have radiation necrosis (Figure 1).7 These data have led some clinicians to suggest that a minimum of 3 cycles of adjuvant temozolomide be given before a conclusion of tumor progression is made.
Several imaging modalities, such as magnetic resonance perfusion with or without spectroscopy and positron emission tomography, are used to help distinguish tumor recurrence from pseudoprogression but are not always reliable.8 In these cases, repeat imaging can be useful, although surgery may be necessary to relieve mass effect and obtain a tissue diagnosis.
Approved therapies for recurrent glioblastoma
There are currently three US Food and Drug Administration (FDA)–approved therapies for recurrent GB: Gliadel wafers; bevacizumab (Avastin), a humanized monoclonal antibody that sequesters vascular endothelial growth factor-A (VEGF); and the NovoTTF-100A System (NovoCure; Portsmouth, NH), which delivers low-intensity, alternating electrical fields (called tumor treatment fields) to the tumor bed that may inhibit cell growth by disrupting microtubule formation.9
The prospective randomized study that led to the approval of Gliadel demonstrated a modest increase in overall survival, from 23 weeks in those patients who received placebo wafers to 31 weeks for those receiving Gliadel.4 This study included recurrent anaplastic gliomas and GB, and the benefit in the GB subgroup was smaller than in the group as a whole. Furthermore, this and other studies predated the use of chemoradiation and adjuvant temozolomide, and it is unclear what benefit, if any, Gliadel offers in recurrent GB patients treated with prior temozolomide.
The approval of bevacizumab for recurrent GB in the United States was based on two clinical trials that evaluated bevacizumab as a single agent or combined with irinotecan in patients with tumor recurrence after initial treatment with chemoradiation and adjuvant temozolomide. The comparative study enrolled 167 patients, 85 in the bevacizumab-alone arm and 82 in the bevacizumab plus irinotecan arm.10 The objective response rate was 25.9% in patients who received bevacizumab monotherapy. There were no complete responses per outside review. The median duration of response was 4.2 months, and the 6-month progression-free survival (PFS-6) was 36.0 %. The single-arm study enrolled 56 patients with recurrent high-grade glioma.11 Objective response as determined by independent review was 19.6%. The median duration of response was 3.9 months.
The FDA approved bevacizumab for use as a single agent based on the improvement in objective response rate in these studies (albeit without improvement in disease-related symptoms or increased survival) and on the fact that patients receiving bevacizumab as a single agent had less toxicity than those receiving the combination with irinotecan and similar outcomes. Subsequent studies with bevacizumab as a single agent in recurrent GB have shown similar PFS-6 rates of between 29% and 42%; smaller studies in recurrent anaplastic gliomas reported response rates ranging between 34% and 68% and PFS-6 rates ranging between 32% and 68%.12,13
The NovoTTF-100A System received approval in the spring of 2011 based on the results of a single multinational study of 237 patients with recurrent GB.14 Patients were randomized to receive either the NovoTTF-100A System or chemotherapy of their physician’s choice. The NovoTTF device includes a battery pack (weighing about 6 pounds) and electrodes, which are placed on the scalp and designed to be worn for about 20 hours a day. The results showed that patients who used the NovoTTF-100A System had higher objective response rates (12% for NovoTTF compared with 6% in the chemotherapy group) and a favorable overall survival rate of 6.6 months compared with 6.0 months for the chemotherapy group. The device was well tolerated, with a significantly higher incidence of toxicities (hematologic and other) in the patients receiving chemotherapy.
Temozolomide is approved in Europe for recurrent high-grade gliomas, including both GB and anaplastic astrocytoma, but in the United States, it is only approved for recurrent anaplastic astrocytoma. These approvals were based on studies in mostly chemotherapy-naive patients, and although it is less clear that rechallenge with temozolomide is useful, as noted below, ongoing studies may clarify this issue.
Approach to the patient with recurrent GB
In the absence of enrollment in a clinical trial, which is encouraged for all glioma patients, the approach to the patient with recurrent GB should take into consideration prior therapies, extent of recurrence and location, and general medical condition. An initial consideration is whether the patient is a candidate for further resection and/or radiation. Re-resection can relieve mass effect and reduce the need for steroids, can provide histologic confirmation of the diagnosis, and likely can increase survival, although this has not been shown in randomized studies.15 For a patient with contraindications for further systemic chemotherapy and who is undergoing resection, Gliadel is a reasonable consideration, and it does not preclude use of bevacizumab or NovoTTF.
Re-irradiation with single-fraction or fractionated stereotactic radiation is feasible in patients with localized recurrent disease. Small, single-arm prospective studies and retrospective reviews suggest a benefit, and the focused delivery modalities reduce the dose to surrounding brain tissue, minimizing the risk of radiation toxicity.16 In some studies, low-dose temozolomide or bevacizumab was combined with re-irradiation with results suggesting both tolerability and efficacy.17 We have found re-irradiation to be a good alternative for those patients who develop profound and prolonged myelosuppression (usually thrombocytopenia) during initial treatment with temozolomide and who have tumor progression before recovery of the counts.
Bevacizumab is currently the most common single agent used for glioma recurrence and is usually dosed at 10 mg/kg every 2 weeks until disease progression, unacceptable toxicity, or the decision to discontinue care. Due to its potent anti-VEGF activity, which results in normalization of highly permeable tumor vessels, bevacizumab often produces rapid and marked reduction in edema and contrast enhancement on neuroimaging.18 This can produce rapid clinical and imaging responses, although whether this reflects true antiglioma activity remains under debate.
Resolution of mass effect and contrast enhancement can also coexist with progression of nonenhancing fluid-attenuated recovery (FLAIR) abnormality that reflects a phenotypically invasive tumor recurrence pattern, with GB co-option of normal cerebral vessels and diffuse, multilobar perivascular spread of tumor cells. The imaging changes can make evaluation of tumor response and progression difficult if one relies on the standard criteria of two-dimensional measurement of enhancing disease; new criteria that take into consideration nonenhancing signal abnormality changes have been proposed.19
Bevacizumab is well tolerated in the brain tumor population, with the same spectrum of side effects seen in other cancer populations (hemorrhage, thrombosis, hypertension, bowel perforation, impaired wound healing, and proteinuria).12,13 The incidence of life-threatening events, such as significant intracranial hemorrhage (3%) or thromboembolism (2%–12%), is within the expected range for the population and is not clearly increased by ¬bevacizumab.12
Several small series have reported that corticosteroid reductions were feasible in 33%–59% of patients with recurrent GB after bevacizumab treatment, and others have reported average corticosteroid dose reductions of 72% and 59%.11,13 This is an important benefit of bevacizumab, as chronic or high-dose corticosteroid use in patients with glioma is associated with significant morbidity. The ability of bevacizumab to control edema confounds the definition of tumor progression. Figure 2 shows a patient who had early neurologic improvement after initiation of bevacizumab and then remained clinically stable without corticosteroids over 14 months of therapy, despite slow continuous growth of the tumor mass.
Bevacizumab is also tolerated by older patients; there is an intriguing study suggesting that not only do older patients tolerate bevacizumab, they may also have increased benefit over younger patients (age separation: younger than 55 years or 55 years and older).20
Another option for some patients, prior to bevacizumab, is rechallenge with temozolomide at alternative dosing schedules, which result in prolonged exposure to higher cumulative doses than that achieved by standard 5-day dosing.21 Resistance to temozolomide occurs through direct repair of DNA by MGMT; a proposed mechanism to overcome resistance would be to deplete tumor-cell MGMT. Several studies have shown that prolonged exposure of peripheral blood mononuclear cells results in depletion of MGMT, and it has been suggested that this could also occur in glioma cells.22 Other studies suggest that prolonged exposure to temozolomide may be directly toxic to endothelial cells.23 These data provide a rationale for temozolomide rechallenge using alternative dose and dosing schedules that deliver higher culmulative doses over prolonged periods.
Commonly tried temozolomide schedules have been 21 days on/7 days off at doses of 75–100 mg/m2, 7 days on/7 days off at a dose of 150 mg/m2, and continuous daily dosing at 50 mg/m2 (Table 1). These schedules were well tolerated in these pretreated patients, with cumulative leukopenia after several cycles. Results have shown PFS-6 of 23%–48% with a suggestion (not supported by all studies) that best responses are seen in patients who were rechallenged after a treatment-free interval (from standard adjuvant temozolomide).24–27 Responses were also similar in patients with high and low levels of tumor MGMT, suggesting that these regimens may overcome -MGMT-mediated resistance.28
In addition to bevacizumab and temozolomide, lomustine, carmustine, irinotecan, cisplatin, and carboplatin have shown modest efficacy in studies as single agents or in combination regimens.29–31 The populations in these studies usually included both recurrent GB and anaplastic tumors, including oligodendrogliomas, and were carried out prior to standard use of chemoradiation and adjuvant temozolomide. Thus, it is difficult to extrapolate how these results would translate into today’s patient population. Interestingly, in a recent randomized phase III trial of recurrent GB (after prior temozolomide), lomustine was found to be superior to the investigational pan--VEFG receptor inhibitor cediranib.32
As noted, the NovoTTF-100A System was only recently approved, and experience is limited. Despite the need for patients to wear the device for 20 hours a day, and for intermittent adjustments to electrode placement at a clinic site, patient compliance in the study was good, and toxicities were minimal. It is clearly an option for patients with recurrent GB, but additional experience is needed to clarify the optimal time of use. The device is currently under study for the treatment of newly diagnosed GB; results of this study may help clarify the role of NovoTTF in treating this ¬malignancy.
In deciding which of the above strategies to use at first or even subsequent recurrences, we take into consideration the rapidity of tumor growth, extent of edema, mass effect, need for corticosteroids, and symptoms. Furthermore, to date there are no agents that improve outcomes when combined with bevacizumab or used after relapse on bevacizumab. This is often the last treatment regimen many patients receive before palliative end-of-life care. Thus, for a patient with a small, asymptomatic recurrence found on surveillance imaging and in the absence of an available clinical trial, we initiate therapy with any of the above-mentioned standard chemotherapeutic agents. At tumor progression and if chemotherapy is still tolerated, another agent may be tried. In contrast, the patient with a rapidly growing, large, or symptomatic recurrence requiring increasing doses of steroids will usually have an immediate clinical benefit from bevacizumab, which will improve quality of life.
Recurrent anaplastic gliomas
There is less consensus on how to treat anaplastic gliomas (anaplastic as¬tro¬cytoma, anaplastic oligodendroglioma, and mixed anaplastic oligoastrocytoma) at initial diagnosis and therefore even more variability in how these patients are treated at recurrence. The study that showed the benefit of chemoradiation and adjuvant temozolomide for newly diagnosed GB excluded patients with anaplastic tumors.1 A meta-analysis of clinical trials included adults with high-grade glioma who after initial surgery were treated with radiation plus chemotherapy (most often a nitrosourea) or radiation. The results only suggested that chemotherapy provided an additional survival benefit over radiotherapy alone for both GB and anaplastic patients.
Many cite the above data as the rationale for including temozolomide in the initial treatment of anaplastic tumors.33 Others, however, cite the results of NOA-04, a large study of patients with anaplastic glioma randomized to receive initial therapy with radiation or one of two chemotherapy regimens: procarbazine (Matulane), lomustine, and vincristine (PCV) or temozolomide.34 At tumor progression, patients who had received radiation were treated with either PCV or temozolomide, and those initially treated with either chemotherapy regimen were irradiated. This study demonstrated no difference in time to treatment failure or PFS among the three groups or any significant difference between the two chemotherapy regimens. Patients with anaplastic astrocytomas fared worse than those with anaplastic oligodendrogliomas or mixed tumors, suggesting that the latter groups may do well with initial chemotherapy only and then radiation at recurrence.
For patients with anaplastic tumors who have not previously been treated with temozolomide, studies do suggest that its use at recurrence is beneficial. In one phase II study, temozolomide-naive patients, or those who had previously received a nitrosourea, showed a 35% overall response rate and PFS-6 of 46% when treated with temozolomide at first recurrence.35 A recent study (RESCUE) evaluating a continuous low-dose temozolomide regimen of 50 mg/m2 in recurrent GB and anaplastic tumors demonstrated a PFS-6 of 35.7% for the anaplastic subgroup that contained patients who had previously had a variety of initial therapies, including chemoradiation.25
With the knowledge that the presence of chromosome 1p/19q codeletions in anaplastic tumors is prognostic for better outcomes,36 two ongoing studies will hopefully provide definitive answers for the treatment of recurrent anaplastic gliomas. The Chemoradiation and Adjuvant Temozolomide in Non-deleted Anaplastic Tumors (CATNON) study will randomly assign patients after surgery to receive either chemoradiation or radiation alone. Following this therapy, there is a second randomization to adjuvant temozolomide or observation only. The trial endpoint is overall survival. The phase III intergroup study of radiotherapy versus temozolomide alone versus radiotherapy with concomitant and adjuvant temozolomide for patients with 1p/19q codeleted anaplastic glioma (CODEL) will determine whether these patients with inherently better outcomes may do just as well with less aggressive therapy.
Several studies of bevacizumab have included recurrent anaplastic gliomas. Two studies of the combination of bevacizumab and irinotecan produced response rates of 55%–66% and PFS-6 of 56%–61%, suggesting activity.12,37 Extrapolating from the GB data, it is likely that single-agent bevacizumab would be efficacious and less toxic than this combination.
Emerging strategies
The identification of prognostic and predictive markers is paving the way for individualized treatment planning. In addition to the prognostic value of 1p/19q codeletions in anaplastic gliomas, the presence of MGMT promoter methylation in GB is likely predictive of response to temozolomide, although this is still under debate. There has been recent excitement about the demonstration that the presence of mutated isocitrate dehydrogenase 1 (IDH1) in gliomas is a robust independent factor associated with better outcome.38 For example, in a series of patients with anaplastic glioma, patients with the IDH1 mutation had a median survival four times longer than that of those without the mutation (81.1 months vs 19.4 months).39 This raises the question of the role of mutated IDH1 in glioma biology and makes it a potentially valuable therapeutic target.
Ongoing gene-expression profiling studies are showing that histologically indistinguishable GB can be clustered into distinct molecular subtypes, with widely different outcomes and responses to treatment.40 This likely contributed to the failure of past clinical trials, as the populations under study were, in fact, too diverse; potentially efficacious agents for one or more subtypes may have been overlooked. These studies are also identifying novel cellular targets such as MET, fibroblast growth factor receptor (FGFR), heat shock protein-90 (HSP-90), and hypoxia-inducible factor 1? (HIF1?).41 Other research is focusing on targets involved in glioma migration and invasion such as tenascin, the Src family of nonreceptor tyrosine kinases, the Rho family of small GTPases, and integrins. The role of glioma stem cells in glioma development and resistance to therapy is another emerging area of study and has led to the identification of specific glioma stem cell targets such as Notch and Sonic hedgehog.42
Conclusion
High-grade gliomas are challenging to treat, and there is often no clear standard of care. The Glioma Outcomes Project tracked clinical practice patterns and outcomes among North American patients with malignant glioma between 1997 and 2000.43 The results showed that patients treated at academic centers were significantly more likely to receive chemotherapy or radiation therapy, to participate in clinical trials, and to have longer survival times than those treated at community centers. Whether these results would be the same today, with the routine use of temozolomide and bevacizumab, is unclear, but they do support referral of these patients to centers with multispecialty clinics. This is, however, not always feasible, and patients may choose to stay close to home.
For patients with recurrent high-grade gliomas, there are several available therapeutic options, including operation, irradiation, and additional systemic therapies, which are available at most centers. Although the optimal sequence in which therapies should be given has not been clarified, these treatments can delay the onset of neurologic deficits and result in improved quality of life and likely prolonged survival. Additionally, the appropriate management of comorbidities such as seizures and brain edema is essential, and several pertinent reviews are available.44,45
References
- Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–996.
- Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997–1003.
- Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009;10:459–466.
- Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-Brain Tumor Treatment Group. Lancet 1995;345:1008–1012.
- Westphal M, Ram Z, Riddle V, et al. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien) 2006;148:269–275.
- Cairncross G, MacDonald D, Ludwin S, et al. Chemotherapy for anaplastic oligodendroglioma. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1994;12:2013–2021.
- Chamberlain MC, Glantz MJ, Chalmers L, et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol 2007;82:81–83.
- Clarke JL, Chang S. Pseudoprogression and pseudoresponse: challenges in brain tumor imaging. Curr Neurol Neurosci Rep 2009;9:241–246.
- Kirson ED, Dbaly V, Tovarys F, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A 2007;104:10152–10157.
- Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 2009;27:4733–4740.
- Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 2009;27:740–745.
- Vredenburgh JJ, Desjardins A, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 2007;25:4722–4729.
- Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology 2008;70:779–787.
- Stupp R, Kanner A, Engelhard H, et al. A prospective, randomized, open-label, phase III clinical trial of NovoTTF-100A versus best standard of care chemotherapy in patients with recurrent glioblastoma. J Clin Oncol 2010;28(18S):LBA2007.
- Park JK, Hodges T, Arko L, et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J Clin Oncol 2010;28:3838–3843.
- Combs SE, Thilmann C, Edler L, et al. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol 2005;23:8863–8869.
- Combs SE, Wagner J, Bischof M, et al. Radiochemotherapy in patients with primary glioblastoma comparing two temozolomide dose regimens. Int J Radiat Oncol Biol Phys 2008;71:999–1005.
- Pope WB, Lai A, Nghiemphu P, et al. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology 2006;66:1258–1260.
- Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:1963–1972.
- Nghiemphu PL, Liu W, Lee Y, et al. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology 2009;72:1217–1222.
- Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro Oncol 2009;11:69–79.
- Spiro TP, Liu L, Majka S, et al. Temozolomide: the effect of once- and twice-a-day dosing on tumor tissue levels of the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase. Clin Cancer Res 2001;7:2309–2317.
- Kurzen H, Schmitt S, Naher H, Mohler T. Inhibition of angiogenesis by non-toxic doses of temozolomide. Anticancer Drugs 2003;14:515–522.
- Wick A, Pascher C, Wick W, et al. Rechallenge with temozolomide in patients with recurrent gliomas. J Neurol 2009;256:734–741.
- Perry JR, Belanger K, Mason WP, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol 2010;28:2051–2057.
- Wick W, Steinbach JP, Kuker WM, Dichgans J, Bamberg M, Weller M. One week on/one week off: a novel active regimen of temozolomide for recurrent glioblastoma. Neurology 2004;62:2113–2115.
- Brandes AA, Tosoni A, Cavallo G, et al. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: phase II study from Gruppo Italiano Cooperativo di Neuro-oncologia (GICNO). Br J Cancer 2006;95:1155–1160.
- Wick A, Felsberg J, Steinbach JP, et al. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol 2007;25:3357–3361.
- Newton HB, Page MA, Junck L, et al. Intra-arterial cisplatin for the treatment of malignant gliomas. J Neurooncol 1989;7:39–45.
- Yung WK, Mechtler L, Gleason MJ. Intravenous carboplatin for recurrent malignant glioma: a phase II study. J Clin Oncol 1991;9:860–864.
- Gwak HS, Youn SM, Kwon AH, Lee SH, Kim JH, Rhee CH. ACNU-cisplatin continuous infusion chemotherapy as salvage therapy for recurrent glioblastomas: phase II study. J Neurooncol 2005;75:173–180.
- Batchelor T, Mulholland P, Neyns B, et al. The efficacy of cediranib as monotherapy and in combination with lomustine compared to lomustine alone in patients with recurrent glioblastoma: a phase III randomized study. J Neurooncol 2010;12(suppl 4):iv69–iv75.
- Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002;359:1011–1018.
- Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol 2009;27:5874–5880.
- Yung WK, Prados MD, Yaya-Tur R, et al. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group. J Clin Oncol 1999;17:2762–2771.
- Jenkins RB, Blair H, Ballman KV, et al. A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 2006;66:9852–9861.
- Desjardins A, Reardon DA, Herndon JE, et al. Bevacizumab plus irinotecan in recurrent WHO grade 3 malignant gliomas. Clin Cancer Res 2008;14:7068–7073.
- Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–1812.
- Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol 2009;27:4150–4154.
- Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17:98–110.
- Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res 2009;29:5171–5184.
- Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY, Olson JJ. Exciting new advances in neuro-oncology: the avenue to a cure for malignant glioma. CA Cancer J Clin 2010;60:166–193.
- Chang SM, Parney IF, Huang W, et al. Patterns of care for adults with newly diagnosed malignant glioma. JAMA 2005;293:557–564.
- Pruitt AA. Treatment of medical complications in patients with brain tumors. Curr Treat Options Neurol 2005;7:323–336.
- Drappatz J, Schiff D, Kesari S, Norden AD, Wen PY. Medical management of brain tumor patients. Neurol Clin 2007;25:1035–1071, ix.
ABOUT THE AUTHORS
Affiliations: Dr. Rosenfeld is Adjunct Professor of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA; Ms. Albright is a Certified Registerd Nurse Practitioner at the University of Pennsylvania School of Medicine; and Dr. Pruitt is Associate Professor of Neurology at the University of Pennsylvania School of Medicine.
Conflicts of interest: The authors have nothing to disclose.
FIGURE 1 Pseudoprogression after chemoradiation. MRI of a biopsy-proven grade III astrocytoma before treatment (left panel). Three weeks after completion of chemoradiation, there is an increase in the size of the cystic component and enhancement (middle panel), which could be interpreted as tumor progression. After 4 cycles of post-radiation temozolomide, the right panel shows a decrease in size and enhancement, supporting the earlier imaging changes as reflective of pseudoprogression. All images are T1 post gadolinium with a 1.5 Tesla magnet.
FIGURE 2 Imaging changes with bevacizumab. This 57-year-old woman presented with a second recurrence of glioblastoma 2 years after chemoradiation and 6 cycles of adjuvant temozolomide and 6 months after 8 cycles of low-dose temozolomide. The images show post-contrast T1-weighted sequences (A, C, E) and fluid-attenuated recovery (FLAIR) sequences (B, D, F). Baseline images (A, B) show a left frontal mass with enhancement, edema, and some mass effect. Eight months later, after 4 cycles of bevacizumab (dosed every 2 weeks with 3 doses per cycle), there is decreased periventricular enhancement and edema (C) and improved FLAIR signal (D) but enlargement of the enhancing left frontal mass, which would meet standard definitions of tumor progression. The patient was neurologically stable and remained on treatment. Nine months later, after 5 additional cycles, she remained neurologically stable (and was not receiving corticosteroids). At this time, the mass has continued to increase in size (note: there is also invasion of the frontal sinus; E), with increasing FLAIR abnormality, likely reflecting a progressive infiltrating nonenhancing tumor (F).
Management of hyperuricemia in adults with or at risk of tumor lysis syndrome
Manuscript received September 9, 2010; accepted April 8, 2011.
* Current affiliation: Incyte Corporation, Wilmington, DE.
Correspondence to: Karen P. Seiter, MD, New York Medical College, Munger Pavilion, Room 250, Valhalla, NY 10595; telephone: 914-493-7514; fax: 914-594-4420; e-mail: [email protected].
Tumor lysis syndrome (TLS) is a relatively common, potentially life-threatening complication of aggressive cytotoxic therapy characterized by metabolite and electrolyte abnormalities (eg, hyperuricemia). To increase the awareness of the risk of hyperuricemia and TLS in adult patients with cancer, who are likely to have age- or lifestyle-related comorbidities, the authors examine the pathophysiology and risk of TLS in adult patients with a broad spectrum of cancer diagnoses. Current recommendations for effective prophylaxis and management of TLS are summarized briefly. Particular emphasis is given to the appropriate role of antihyperuricemic therapy with rasburicase in adults, based on the recent results of a phase III clinical study.
Tumor lysis syndrome (TLS) is a serious, potentially life-threatening condition of metabolic derangement and impaired electrolyte homeostasis. TLS can occur in patients with cancer as a result of spontaneous or, more commonly, treatment-induced tumor cell death and typically is seen in patients with hematologic, rather than solid organ, malignancies who are undergoing chemotherapy. It is particularly an issue for patients with rapidly growing tumors and a high tumor burden (as evidenced by a high white blood cell [WBC] count in leukemia and elevated serum lactate dehydrogenase [LDH] levels and/or advanced clinical stage in lymphoma) at the beginning of chemotherapy. TLS is caused by a sudden, massive release of cell content from lysed tumor cells into the bloodstream, which overwhelms the body’s capacity for homeostatic regulation. Because persistent or progressive metabolic derangement is associated with a high risk of organ failure, TLS is a clinical emergency.
To facilitate the diagnosis, prevention, and treatment of TLS, Cairo and Bishop established precise criteria for the categorization of TLS on the basis of metabolic abnormalities and the associated significant clinical toxicities that require clinical intervention.1 According to Cairo and Bishop, laboratory TLS (LTLS) is defined by the presence of two or more of the following metabolic abnormalities: hyperuricemia, hyperkalemia, hyperphosphatemia, and secondary hypocalcemia. Clinical TLS (CTLS) is defined as LTLS accompanied by at least one clinical complication, such as renal impairment (defined as a serum creatinine concentration greater than 1.5 times the upper limit of normal), cardiac arrhythmia, or seizure. The aforementioned clinical manifestations should not be directly or likely attributable to a therapeutic agent (eg, a rise in creatinine levels after administration of a nephrotoxic drug).1
Estimates from clinical studies of the incidence of TLS vary widely.2 In a review of case records of 102 patients with high-grade non-Hodgkin’s lymphoma (NHL), 42% of patients had signs of LTLS and 6% had CTLS.3 In contrast, analysis of data from a sample of 755 European children and adults with newly diagnosed or recurrent acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), or NHL indicated a 19% incidence of hyperuricemia and a 5% incidence of LTLS per episode of administration of an induction therapy ¬regimen.4
Pathophysiology of TLS in adults
The pathophysiology and clinical consequences of TLS have been discussed infrequently in the context of adult patient populations. Yet adult ¬patients are more likely than pediatric patients to experience potentially serious metabolic, cardiac, renal, or multisystemic comorbidities. Such comorbidities, whether chronic illnesses present at the initiation of anticancer therapy or acute conditions that develop during administration of aggressive cytoreductive regimens, require special consideration because they may amplify the metabolic and electrolyte imbalances caused by tumor cell lysis and may have secondary pathophysiologic consequences. In adults, these effects may further weaken an already strained homeostatic regulation and thereby significantly increase the risk of serious clinical complications of TLS.5 Elderly patients (age > 65 years) with cancer are particularly likely to have comorbidities, which may worsen their prognosis in the event of TLS, including baseline chronic renal insufficiency and/or heart disease.6
Hyperuricemia is one of the hallmarks of LTLS. If not reversed quickly, severe hyperuricemia may have serious clinical consequences, particularly acute kidney injury (AKI), which is an independent risk factor for mortality (even mild AKI).2 Additionally, hyperuricemia can lead to a variety of distressing symptoms, such as gastrointestinal complaints (nausea, vomiting, diarrhea, and anorexia), lethargy, hematuria, flank or back pain, fluid overload, edema, arthralgias, hypertension, and signs of obstructive uropathy.7
The risk of renal complications is particularly high in elderly patients with baseline renal disease, which may have been caused by diabetes, hypertension, renal artery stenosis, chronic pyelonephritis, amyloidosis, glomerular disease, or treated malignancies.6 Furthermore, congestive heart failure, use of thiazide or loop diuretics, obesity, type II diabetes, renal impairment, hypertriglyceridemia, and peripheral vascular disease are major cardiovascular risk factors associated with hyperuricemia.6
Serious clinical consequences may arise not only from hyperuricemia but also from TLS-related electrolyte abnormalities (ie, hyperphosphatemia, hyperkalemia, or hypocalcemia). AKI, cardiac arrhythmia, and neurologic impairment, such as seizures or higher CNS dysfunction, may result from severe hyperphosphatemia and secondary hypocalcemia caused by calcium phosphate precipitation in renal tubules. These two metabolic disturbances also may present with muscle cramps, tetany, or perioral numbness or tingling; rare symptoms include fatigue, bone and joint pain, pruritus, and rash.7 Hyperphosphatemia can be exacerbated by excessive use of phosphate-containing laxatives or enemas, which is especially common in the elderly. Hyperkalemia also can lead to neuromuscular symptoms, but principally, it may cause potentially life-threatening cardiac dysfunction.2
Because of the decrease in physiologic reserves with age, the aforementioned electrolyte abnormalities are associated with increased morbidity and mortality in the elderly.8 For example, renal function generally declines with age, and age-related reduction in renin and aldosterone levels increases the risk of hyperkalemia.6 Particularly among the elderly, the risk of hyperkalemia is increased in those with renal tubular reabsorption/secretion defects, those with type II diabetes who develop type IV renal tubular acidosis as the result of hyporeninemic hypoaldosteronism, and those who take nonsteroidal anti-inflammatory drugs on a long-term, scheduled basis.9,10 Although antihypertensive medications generally have cardioprotective and renoprotective properties, many antihypertensives actually may increase the risk of hyperkalemia in elderly patients with renal tubular acidosis.11,12 Also in adults, hypocalcemia may result from vitamin D deficiency, impaired vitamin D metabolism, low intestinal Ca2+ absorption, phosphate retention, chronic hypomagnesemia, serum protein abnormalities, and parathyroid hormone resistance; it may also occur as a medication adverse effect (specifically relevant for patients with cancer treated with bisphosphonates, anticonvulsants, cis¬platin, and the combination of 5-fluor¬¬ouracil and ¬leucovorin).6,13
Risk of TLS in adults with cancer
The risk of TLS in adults with cancer depends on multiple components, including disease-related factors, the type and aggressiveness of anticancer treatment, other medications concomitantly administered, and patient (host)-related factors. TLS has been observed primarily in patients with hematologic malignancies, with particularly high incidences reported for those with Burkitt (and Burkitt-like) lymphoma, precursor B-lymphoblastic leukemia/lymphoma, high-stage T-cell anaplastic large cell lymphoma, and ALL.2
Demographic data from compassionate-use studies of the uricolytic agent rasburicase (Elitek) in patients with or at high risk of acute cancer-associated hyperuricemia and TLS seem to suggest that AML with high WBC counts and select types of NHL (mostly high-grade and some intermediate-grade lymphomas) are associated with a high risk of TLS in adults.14,15 Based on the findings of two European studies, between 3% and 17% of adult patients with AML may experience LTLS or CTLS in response to induction therapy.4,16 In one of the two studies, approximately 20% of adults with AML, ALL, or NHL experienced hyperuricemia after induction therapy.4 Data from the second study, which included 772 adults with AML (including some cases of chronic myelogenous leukemia [CML] in blast crisis) who had received induction chemotherapy, were used to develop a risk-prediction model for CTLS in adult patients with AML.16 According to the model, high WBC count, pretreatment hyperuricemia, and high baseline serum creatinine and LDH concentrations were significant independent prognostic factors for the development of LTLS and CTLS.16
An independent international panel of experts in pediatric and adult hematologic malignancies and TLS recently developed guidelines for the management of TLS based on a comprehensive risk-stratification algorithm. These guidelines were published in 2008 by the American Society of Clinical Oncology.17 Patients were considered to be at high, intermediate, or low risk of TLS, depending on the specific type of malignancy, tumor burden, and type of cytoreductive therapy administered.
In addition to these basic risk categories, other risk factors such as renal function and plasma uric acid (PUA) level at baseline were incorporated into the recommendations for TLS prevention and treatment.17 For example, the presence of baseline hyperuricemia (defined as a serum uric acid level > 7.5 mg/dL) is a modifier of the recommendation of antihyperuricemic therapy for patients at intermediate risk of TLS; if there are high baseline PUA levels, the drug of choice should not be allopurinol but rather rasburicase.17 However, these guidelines do not address all malignancies or uniformly assess risk depending on renal involvement by the disease or kidney function.
Consequently, another consensus panel was convened to build upon the 2008 guidelines and produce a medical decision tree for ranking patients with cancer as low, intermediate, or high risk of TLS. For this project, risk factors included biologic evidence of LTLS, tumor proliferation, and bulk and stage of malignant tumor, as well as renal impairment and/or involvement by the disease at the time of TLS diagnosis; subsequently, an algorithmic model of low-, intermediate-, and high-risk TLS classification and associated TLS prophylaxis recommendations were finalized.18 TLS risk factors of particular relevance for elderly patients are age-related alterations in heart anatomy and function, obesity, generalized deconditioning, alterations of the cardiovascular and circulatory systems, use of multiple drugs with potential pharmacodynamic interactions, age-related decrease in glomerular filtration rate, tobacco use, excessive alcohol consumption, and unhealthy dietary ¬habits.6
In addition to hematologic malignancies, solid tumors may result in TLS as a response to chemo(bio)-therapy or radiation/chemoradiation therapy or spontaneously. In adults, solid tumors that have been associated with TLS include, but are not limited to, breast cancer, lung cancer (especially small cell lung cancer), gastrointestinal stromal tumor, germ cell tumor, hepatocellular carcinoma, melanoma, malignant pheochromocytoma, ovarian cancer, colon cancer, and renal cell carcinoma.19–21 Table 1 presents a list of recent reports of TLS in adults with hematologic malignancies or solid tumors.22–41 Although TLS is less common in patients with solid tumors than in those with hematologic malignancies, its onset and progression in patients with solid tumors often are less predictable.19 Consequently, patients with TLS from solid tumors tend to have worse prognoses.19 In patients with solid tumors, TLS may develop days or even weeks after initiation of chemotherapy, providing no or limited opportunity for effective prophylaxis. Thus, for patients with solid tumors, heightened awareness of and vigilance for TLS are necessary to ensure appropriate and timely management of TLS.
The importance of such timely management of TLS is illustrated by several case studies. In one such case, a 55-year-old man with advanced hepatocellular carcinoma developed acute renal failure, hyperkalemia, and hyperuricemia 30 days after the initiation of treatment with the oral tyrosine kinase inhibitor sorafenib (Nexavar). The patient eventually died of multiple organ failure, despite treatment with hyperhydration, administration of the maximal daily dose of allopurinol, and use of emergency hemodialysis.37
Another fatal outcome of TLS was reported for a 62-year-old man with metastatic colon cancer and a 10-year complicated history of treatments. The patient, who had extensive lung metastases, hyperuricemia, and elevated serum LDH and creatinine concentrations at baseline, developed severe TLS within 2 days of receiving bevacizumab (Avastin) with combination chemotherapy and eventually died of acute renal failure.34
Furthermore, a rare case of spontaneous TLS occurred in a patient with Crohn’s disease who developed plasmacytoma while being treated with immunosuppressants. The patient experienced extreme hyperuricemia (a PUA level of 44 mg/dL) and died of the consequences of acute oliguric renal failure, despite hyperhydration, alkalinization, treatment with maximal doses of allopurinol followed by rasburicase, and hemodialysis.39
According to current TLS management guidelines, patients with multiple myeloma or rapidly growing solid tumors and an expected rapid response to therapy are considered to be at intermediate risk of TLS. However, the risk of TLS in patients with solid tumors is increased by the presence of bulky disease (masses or lymph nodes > 10 cm),17 unfavorable host baseline characteristics (such as renal insufficiency and baseline hyperuricemia),17 and the presence of liver metastases.19 According to relatively recent case reports, TLS occurred in two adult patients with hepatocellular carcinoma undergoing transarterial chemoembolization36 and in a 58-year-old man with metastatic melanoma and bulky liver metastases, who developed TLS within 24 hours after receiving intra-arterial infusion of cisplatin and embolization therapy.42 TLS from palliative radiotherapy, although rare, also has been reported; two fatal cases include a 74-year-old man with diffuse large B-cell lymphoma43 and a 52-year-old man with non-small cell lung cancer.38
Chemo(bio)therapy regimens associated with risk of TLS
The availability of new agents (either as monotherapy or in combination regimens) with high tumor response rates is expected to increase the risk and incidence of TLS. Even for hematologic malignancies with a relatively low incidence of TLS, such as chronic lymphocytic leukemia (CLL), the risk of TLS may increase with the growing use of novel agents that are highly effective inducers of apoptosis.44
In CLL associated with WBC counts of 10,000–100,000 cells/µL, fludarabine therapy (as monotherapy or combination therapy) confers intermediate-risk status (for TLS), according to current TLS management guidelines.17 Current US prescribing information for rituximab (Rituxan), which together with fludara¬bine (± cyclophosphamide) constitutes the standard treatment backbone for previously untreated CLL,45 contains a black-box warning for TLS when rituximab is given for the treatment of NHL.46 A warning for TLS also has been issued for the use of bendamustine (Treanda) for CLL.47 Alvocidib (flavopiridol), a cyclin-dependent kinase inhibitor in clinical development (phase II studies) for the treatment of adults with relapsed CLL, has been associated with a high incidence of TLS, particularly in patients with WBC counts > 200,000 cells/µL.48,49 Finally, lenalidomide (Revlimid) was associated with TLS and a tumor flare phenomenon in patients with CLL in phase II clinical studies.50
A number of case reports have associated imatinib (Gleevec), bortezomib (Velcade), and thalidomide (Thalomid) with the development of TLS in adults. Administration of imatinib led to TLS in two patients treated for ALL and in one patient treated for CML.23 Bortezomib and/or thalidomide caused TLS in several patients treated for multiple myeloma.31,51–54 TLS also is common in patients receiving chemotherapy for acute adult human T-cell lymphotrophic virus-1–associated T-cell leukemia/lymphoma (ATLL). Fatal TLS recently occurred in an obese 57-year-old woman with ATLL, in the background of systemic lupus erythematosus, after chemotherapy with high-dose prednisone and adjusted doses of cyclophosphamide and doxorubicin.33
Management of hyperuricemia in adults: the role of rasburicase
Because hyperuricemia can increase both the risk and the severity of CTLS, management of hyperuricemia that focuses on its prevention and, in cases where it has already emerged, its rapid reversal is an important strategy in reducing TLS-associated mortality and morbidity. The current recommendations for the prevention and management of TLS-associated hyperuricemia have been reviewed in great detail recently.2 Monitoring and clinical judgment generally are adequate for low-risk patients, but patients with higher risk require hydration and antihyperuricemic management with appropriate drug therapy.2 For most patients with an intermediate risk of TLS, prophylactic antihyperuricemic therapy with allopurinol is sufficient, because allopurinol can prevent the buildup of uric acid by blocking the enzymatic conversion of hypoxanthine and xanthine to uric acid (via direct inhibition of xanthine oxidase). However, allopurinol does not modify existing uric acid pools and thus is inadequate for the prevention or management of hyperuricemia in high-risk patients.2 Because a TLS-associated rise in uric acid levels can occur suddenly and rapidly in patients at high risk, effective and safe degradation of uric acid is essential for preventing or reversing life-threatening hyperuricemia.
The effectiveness of rasburicase as antihyperuricemic therapy for adult patients with cancer was demonstrated initially in two international multicenter compassionate-use studies. One study, conducted in the United States and Canada, included 387 adults (and 682 children) with mostly hematologic malignancies who received daily 30-minute IV administrations of rasburicase for 1–7 days at a dose of 0.2 mg/kg.15 All patients who received rasburicase for TLS prophylaxis, including 126 adults, maintained low PUA levels during chemotherapy, and all 212 adults with hyperuricemia at baseline responded to treatment with prompt and impressive reduction in PUA levels, which was maintained for several days.15 Rasburicase generally was well tolerated; only two adults experienced a single episode each of grade 3 toxicity, and no adult experienced grade 4 toxicity.15
The second study—a multicenter compassionate-use study in patients with leukemia or lymphoma from Europe and Australia—included 112 adults (and 166 children) with generally large tumor burdens, as assessed by WBC counts for leukemia and clinical stage and also by serum LDH levels for lymphoma.14 Reduction in PUA levels with rasburicase was statistically and clinically highly significant, resulting in PUA response rates of 100% in both adults and children. Only one adult patient, a 41-year-old man with AML and hyperuricemia at baseline, required hemodialysis because of acute renal failure, although PUA levels returned to normal after treatment with rasburicase. The patient subsequently died of respiratory failure attributable to disease ¬progression.14
The multicenter US registrational trial for the adult indication of rasburicase included 280 patients with hematologic malignancies who were at risk of TLS.55 Patients were randomly assigned to one of three treatments: rasburicase (0.2 mg/kg/d given as a 30-minute IV infusion) for 5 days, rasburicase for 3 days followed by oral allopurinol (300 mg/d) for 2 days, or allopurinol for 5 days. Rasburicase alone achieved significantly greater response rates than allopurinol monotherapy both in patients with a high risk of TLS and in those with baseline hyperuricemia (Table 2). In this clinical trial, control of PUA concentrations was achieved within 4 hours with rasburicase (compared with a median time interval of 27 hours with allopurinol) and was maintained during the 7-day monitoring period after initiation of antihyperuricemic therapy. Although reduction in the incidence of TLS was not an efficacy endpoint of the study, safety data analysis revealed that the occurrence of LTLS or CTLS was less common in the rasburicase than in the allopurinol treatment arm.55
In this large, randomized clinical study, rasburicase was safe and generally well tolerated during the 5-day treatment period. Patients who received rasburicase and those who received allopurinol had a similar incidence of adverse events.55,56 The only drug-related adverse events observed among the 184 patients who received rasburicase (alone or in combination with allopurinol) were potential hypersensitivity reactions in five patients, including irritation at the injection site (one patient); arthralgia, myalgia, and rash (one patient); peripheral edema (one patient); and grade 3 hypersensitivity (two patients). One patient with grade 3 hypersensitivity related to rasburicase discontinued study participation on day 1. No grade 4 hypersensitivity reaction, anaphylaxis, hemolytic reaction, or methemoglobinemia occurred with rasburicase.55
In summary, the results of this phase III randomized controlled study show that rasburicase may be superior to allopurinol in preventing hyperuricemia in adult leukemia/lymphoma patients at high risk of TLS and in achieving rapid and effective control of PUA levels in patients with hyperuricemia. The results support the recommendation within the current TLS management guidelines of using rasburicase for prophylaxis in high-risk patients and for treatment of hyperuricemia of malignancy, as well as in those patients with fully developed LTLS or CTLS (irrespective of PUA levels).17
Conclusion
In adult patients with cancer, the risk of serious clinical consequences of TLS often is increased by the presence of renal impairment, heart disease, baseline metabolic imbalances, and multiorgan derangements. Even in adult patients without serious comorbidities, an age-related decrease in physiologic resistance (reduced pathobiologic reserve), lifestyle factors (eg, adverse chronic exposure to tobacco, alcohol, or other toxins; sedentary existence; obesity; and vitamin and macro/micronutrient deficiencies), and dependence on multiple medications (polypharmacy and an associated increased risk for drug-drug interactions) should be considered for both TLS risk assessment and therapeutic choices in TLS prophylaxis and treatment. Although TLS is most common in patients with hematologic malignancies, serious cases of TLS have been reported in patients with a large variety of solid tumors, with sometimes fatal outcomes.
Assessment of TLS risk in patients with solid tumors, who generally tend to belong to a more aged population, is more difficult than in those with hematologic malignancies. Tumor burden and the type of chemotherapy are the most important risk factors for development of TLS in association with solid tumors; however, an unexpected occurrence of acute spontaneous TLS with hyperuricemia has been observed, even in the absence of these two factors.
Hyperuricemia is a key manifestation of TLS and is associated with risks of AKI, fluid overload, heart failure, and death. Prevention of hyperuricemia in high-risk patients and its rapid reversal in those with hyperuricemia at presentation or in those with fully developed TLS are of utmost importance in the successful management of TLS in adults with cancer. Results of the recent phase III study of rasburicase in adult patients with cancer demonstrated that rasburicase is superior to allopurinol in providing rapid and effective control of PUA levels.
Acknowledgments: Editorial assistance provided by Roland Tacke, PhD, and Candace Lundin, DVM, MS, was funded by sanofi-aventis US. The authors were fully responsible for all content and editorial decisions and did not receive financial support or compensation related to the development of this article.
References
- Cairo MS, Bishop M. Tumour lysis syndrome: new therapeutic strategies and classification. Br J Haematol 2004;127:3–11.
- Mughal TI, Ejaz AA, Foringer JR, Coiffier B. An integrated clinical approach for the identification, prevention, and treatment of tumor lysis syndrome. Cancer Treat Rev 2010;36:164–176.
- Hande KR, Garrow GC. Acute tumor lysis syndrome in patients with high-grade non-Hodgkin’s lymphoma. Am J Med 1993;94:133–139.
- Annemans L, Moeremans K, Lamotte M, et al. Incidence, medical resource utilisation and costs of hyperuricemia and tumour lysis syndrome in patients with acute leukaemia and non-Hodgkin’s lymphoma in four European countries. Leuk Lymphoma 2003;44:77–83.
- Coiffier B, Riouffol C. Management of tumor lysis syndrome in adults. Expert Rev Anticancer Ther 2007;7:233–239.
- Pumo V, Sciacca D, Malaquarnera M. Tumor lysis syndrome in elderly. Crit Rev ¬Oncol Hematol 2007;64:31–42.
- Kennedy LD, Ajiboye VO. Rasburicase for the prevention and treatment of hyperuricemia in tumor lysis syndrome. J Oncol Pharm Pract 2010;16:205–213.
- Allison SP, Lobo DN. Fluid and electrolytes in the elderly. Curr Opin Clin Nutr Metab Care 2004;7:27–33.
- Michelis MF. Hyperkalemia in the elderly. Am J Kidney Dis 1990;16:296–299.
- Williams ME. Endocrine crises: hyperkalemia. Crit Care Clin 1991;7:155–174.
- Weir MR. Non-diuretic-based antihypertensive therapy and potassium homeostasis in elderly patients. Coron Artery Dis 1997;8:499–504.
- Sica DA. Antihypertensive therapy and its effects on potassium homeostasis. J Clin Hypertens (Greenwich) 2006;8:67–73.
- Moe SM. Disorders involving calcium, phosphorus, and magnesium. Prim Care 2008;35:215–237.
- Bosly A, Sonet A, Pinkerton CR, et al. Rasburicase (recombinant urate oxidase) for the management of hyperuricemia in patients with cancer: report of an international compassionate use study. Cancer 2003;98:1048–1054.
- Jeha S, Kantarjian H, Irwin D, et al. Efficacy and safety of rasburicase, a recombinant urate oxidase (Elitek), in the management of malignancy-associated hyperuricemia in pediatric and adult patients: final results of a multicenter compassionate use trial. Leukemia 2005;19:34–38.
- Montesinos P, Lorenzo I, Martin G, et al. Tumor lysis syndrome in patients with acute myeloid leukemia: identification of risk factors and development of a predictive model. Haematologica 2008;93:67–74.
- Coiffier B, Altman A, Pui CH, Younes A, Cairo MS. Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review. J Clin Oncol 2008;26:2767–2778.
- Cairo MS, Coiffier B, Reiter A, Younes A; TLS Expert Panel. Recommendations for the evaluation of risk and prophylaxis of tumour lysis syndrome (TLS) in adults and children with malignant diseases: an expert TLS panel consensus. Br J Haematol 2010;149:578–586.
- Gemici C. Tumour lysis syndrome in solid tumours. Clin Oncol (R Coll Radiol) 2006;18:773–780.
- Pinder EM, Atwal GS, Ayantunde AA, et al. Tumour lysis syndrome occurring in a patient with metastatic gastrointestinal stromal tumour treated with Glivec (imatinib mesylate, Gleevec, STI571). Sarcoma 2007;2007:82012.
- Mott FE, Esana A, Chakmakjian C, Herrington JD. Tumor lysis syndrome in solid tumors. Support Cancer Ther 2005;2:188–191.
- Konuma T, Ooi J, Takahashi S, et al. Fatal acute tumor lysis syndrome following intrathecal chemotherapy for acute lymphoblastic leukemia with meningeal involvement. Intern Med 2008;47:1987–1988.
- Chang H, Shih LY. Imatinib-induced tumor lysis syndrome: report of a case and review of the literature. Chang Gung Med J 2008;31:510–514.
- Rüger W, Kruip MJ, Betjes MG. Reversible renal failure due to bilateral renal sarcoma in a patient with acute myeloid leukemia. Ren Fail 2009;31:606–609.
- Kosseifi SG, Vyas B, Vyas H, Krishnaswamy G, Smith S. Tumor lysis syndrome following chlorambucil therapy for chronic lymphocytic leukemia: case report and review of the literature. Tenn Med 2009;102:33–36.
- Calvo-Villas JM, Urcuyo BM, Umpierrez AM, Sicilia F. Acute tumor lysis syndrome during oral fludarabine treatment for chronic lymphocytic leukemia: role of treatment with rasburicase. Onkologie 2008;31:197–199.
- Al-Kali A, Farooq S, Tfayli A. Tumor lysis syndrome after starting treatment with Gleevec in a patient with chronic myelogenous leukemia. J Clin Pharm Ther 2009;34:607–610.
- Sonmez M, Ovali E, Omay SB. Tumor lysis syndrome during treatment with AMN107 (nilotinib) in a patient with chronic myelogenous leukemia accelerated phase. J Clin Pharm Ther 2008;33:91–92.
- Lin CJ, Chen HH, Hsieh RK, Chen YC, Wu CJ. Acute tumor lysis syndrome in a hemodialysis patient with diffuse large B cell lymphoma. Med Oncol 2009;26:93–95.
- Kasahara I, Nishio M, Yamamoto S, et al. Cord blood transplantation with a reduced-intensity conditioning regimen for patients with relapsed aggressive multiple myeloma after cytoreduction with bortezomib. Int J Hematol 2009;90:413–415.
- Chim CS. Rapid complete remission in multiple myeloma with bortezomib/thalidomide/dexamethasone combination therapy following development of tumor lysis syndrome. Cancer Chemother Pharmacol 2008;62:181–182.
- Bouaziz JD, Cordel N, Hickman G, Fieschi C, Ortonne N, Bagot M. Cutaneous tumor lysis syndrome in a patient with HTLV-1 adult T-cell lymphoma/leukemia. Blood 2009;114:4320–4321.
- Fritsch-Stork RD, Leguit RJ, Derksen RH. Rapidly fatal HTLV-1-associated T-cell leukemia/lymphoma in a patient with SLE. Nat Rev Rheumatol 2009;5:283–287.
- Hentrich M, Schiel X, Scheidt B, Reitmeier M, Hoffmann U, Lutz L. Fatal tumor lysis syndrome after irinotecan/5-FU/folinic acid/bevacizumab-containing therapy in a patient heavily pretreated for metastatic colon cancer. Acta Oncol 2008;47:155–156.
- Feres GA, Salluh JI, Ferreira CG, Soares M. Severe acute tumor lysis syndrome in patients with germ-cell tumors. Indian J Urol 2008;24:555–557.
- Hsieh PM, Hung KC, Chen YS. Tumor lysis syndrome after transarterial chemoembolization of hepatocellular carcinoma: case reports and literature review. World J Gastroenterol 2009;15:4726–4728.
- Huang WS, Yang CH. Sorafenib induced tumor lysis syndrome in an advanced hepatocellular carcinoma patient. World J Gastroenterol 2009;15:4464–4466.
- Noh GY, Choe DH, Kim CH, Lee JC. Fatal tumor lysis syndrome during radiotherapy for non-small-cell lung cancer. J Clin Oncol 2008;26:6005–6006.
- Froilán Torres C, Castro Carbajo P, Pajares Villarroya R, et al. Acute spontaneous tumor lysis syndrome in a patient with Crohn’s disease taking immunosuppressants. Rev Esp Enferm Dig 2009;101:288–294.
- Michels J, Lassau N, Gross-Goupil M, Massard C, Mejean A, Escudier B. Sunitinib inducing tumor lysis syndrome in a patient treated for renal carcinoma. Invest New Drugs 2010;28:690–693.
- Shenoy C. Acute spontaneous tumor lysis syndrome in a patient with squamous cell carcinoma of the lung. QJM 2009;102:71–73.
- Nakamura Y, Nakamura Y, Hori E, et al. Tumor lysis syndrome after transcatheter arterial infusion of cisplatin and embolization therapy for liver metastases of melanoma. Int J Dermatol 2009;48:763–767.
- Yamazaki H, Hanada M, Horiki M, et al. Acute tumor lysis syndrome caused by palliative radiotherapy in patients with diffuse large B-cell lymphoma. Radiat Med 2004;22:52–55.
- Cheson BD. Etiology and management of tumor lysis syndrome in patients with chronic lymphocytic leukemia. Clin Adv Hematol Oncol 2009;7:263–271.
- Lin TS. Novel agents in chronic lymphocytic leukemia: efficacy and tolerability of new therapies. Clin Lymphoma Myeloma 2008;8(suppl 4):S137–S143.
- Rituxan [package insert]. South San Francisco, CA: Genentech, Inc.; 2011.
- Treanda [package insert]. Frazer, PA: Cephalon, Inc.; 2010.
- Lin TS, Ruppert AS, Johnson AJ, et al. Phase II study of flavopiridol in relapsed chronic lymphocytic leukemia demonstrating high response rates in genetically high-risk disease. J Clin Oncol 2009;27:6012–6018.
- Byrd JC, Lin TS, Dalton JT, et al. Flavopiridol administered using a pharmacologically derived schedule is associated with marked clinical efficacy in refractory, genetically high-risk chronic lymphocytic leukemia. Blood 2007;109:399–404.
- Moutouh-de Parseval LA, Weiss L, DeLap RJ, Knight RD, Zeldis JB. Tumor lysis syndrome/tumor flare reaction in lenalidomide-treated chronic lymphocytic leukemia. J Clin Oncol 2007;25:5047.
- Sezer O, Vesole DH, Singhal S, et al. Bortezomib-induced tumor lysis syndrome in multiple myeloma. Clin Lymphoma Myeloma 2006;7:233–235.
- Huston A, Brown J, Roodman GD. Tumor lysis syndrome following thalidomide and dexamethasone therapy for newly diagnosed multiple myeloma. Exp Hematol 2006;34:1616.
- Berenson JR, Yang HH, Vescio RA, et al. Safety and efficacy of bortezomib and melphalan combination in patients with relapsed or refractory multiple myeloma: updated results of a phase 1/2 study after longer follow-up. Ann Hematol 2008;87:623–631.
- Fuente N, Mañe JM, Barcelo R, Muñoz A, Perez-Hoyos T, Lopez-Vivanco G. Tumor lysis syndrome in a multiple myeloma patient treated with thalidomide. Ann Oncol 2004;15:537.
- Cortes J, Moore JO, Maziarz RT, et al. Control of plasma uric acid in adults at risk for tumor lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone: results of a multicenter phase III study. J Clin Oncol 2010;28:4207–4213.
- Elitek [package insert]. Bridgewater, NJ: sanofi-aventis U.S., LLC; 2009.
ABOUT THE AUTHORS
Affiliations: Dr. Seiter is Professor of Medicine and Director of the Leukemia Service at New York Medical College, Valhalla, NY. Dr. Sarlis is currently Vice President and Head of the Medical Affairs Department at Incyte Corporation, Wilmington, DE. Dr. Kim is Associate Professor and Chief, Section of Head and Neck Medical Oncology, Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, at The University of Texas MD Anderson Cancer Center, Houston, TX.
Conflicts of interest: Dr. Sarlis was an employee of sanofi-aventis U.S. at the time this article was written and holds stock options and stock in this company. Drs. Seiter and Kim have no pertinent conflicts of interest to disclose.
Karen P. Seiter, MD,1 Nicholas J. Sarlis, MD, PhD, FACP,2* and Edward S. Kim, MD3
- New York Medical College, Valhalla, NY;
- Medical Affairs–Oncology, sanofi-aventis U.S., Bridgewater, NJ; and
- The University of Texas MD Anderson Cancer Center, Houston, TX
Manuscript received September 9, 2010; accepted April 8, 2011.
* Current affiliation: Incyte Corporation, Wilmington, DE.
Correspondence to: Karen P. Seiter, MD, New York Medical College, Munger Pavilion, Room 250, Valhalla, NY 10595; telephone: 914-493-7514; fax: 914-594-4420; e-mail: [email protected].
Tumor lysis syndrome (TLS) is a relatively common, potentially life-threatening complication of aggressive cytotoxic therapy characterized by metabolite and electrolyte abnormalities (eg, hyperuricemia). To increase the awareness of the risk of hyperuricemia and TLS in adult patients with cancer, who are likely to have age- or lifestyle-related comorbidities, the authors examine the pathophysiology and risk of TLS in adult patients with a broad spectrum of cancer diagnoses. Current recommendations for effective prophylaxis and management of TLS are summarized briefly. Particular emphasis is given to the appropriate role of antihyperuricemic therapy with rasburicase in adults, based on the recent results of a phase III clinical study.
Tumor lysis syndrome (TLS) is a serious, potentially life-threatening condition of metabolic derangement and impaired electrolyte homeostasis. TLS can occur in patients with cancer as a result of spontaneous or, more commonly, treatment-induced tumor cell death and typically is seen in patients with hematologic, rather than solid organ, malignancies who are undergoing chemotherapy. It is particularly an issue for patients with rapidly growing tumors and a high tumor burden (as evidenced by a high white blood cell [WBC] count in leukemia and elevated serum lactate dehydrogenase [LDH] levels and/or advanced clinical stage in lymphoma) at the beginning of chemotherapy. TLS is caused by a sudden, massive release of cell content from lysed tumor cells into the bloodstream, which overwhelms the body’s capacity for homeostatic regulation. Because persistent or progressive metabolic derangement is associated with a high risk of organ failure, TLS is a clinical emergency.
To facilitate the diagnosis, prevention, and treatment of TLS, Cairo and Bishop established precise criteria for the categorization of TLS on the basis of metabolic abnormalities and the associated significant clinical toxicities that require clinical intervention.1 According to Cairo and Bishop, laboratory TLS (LTLS) is defined by the presence of two or more of the following metabolic abnormalities: hyperuricemia, hyperkalemia, hyperphosphatemia, and secondary hypocalcemia. Clinical TLS (CTLS) is defined as LTLS accompanied by at least one clinical complication, such as renal impairment (defined as a serum creatinine concentration greater than 1.5 times the upper limit of normal), cardiac arrhythmia, or seizure. The aforementioned clinical manifestations should not be directly or likely attributable to a therapeutic agent (eg, a rise in creatinine levels after administration of a nephrotoxic drug).1
Estimates from clinical studies of the incidence of TLS vary widely.2 In a review of case records of 102 patients with high-grade non-Hodgkin’s lymphoma (NHL), 42% of patients had signs of LTLS and 6% had CTLS.3 In contrast, analysis of data from a sample of 755 European children and adults with newly diagnosed or recurrent acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), or NHL indicated a 19% incidence of hyperuricemia and a 5% incidence of LTLS per episode of administration of an induction therapy ¬regimen.4
Pathophysiology of TLS in adults
The pathophysiology and clinical consequences of TLS have been discussed infrequently in the context of adult patient populations. Yet adult ¬patients are more likely than pediatric patients to experience potentially serious metabolic, cardiac, renal, or multisystemic comorbidities. Such comorbidities, whether chronic illnesses present at the initiation of anticancer therapy or acute conditions that develop during administration of aggressive cytoreductive regimens, require special consideration because they may amplify the metabolic and electrolyte imbalances caused by tumor cell lysis and may have secondary pathophysiologic consequences. In adults, these effects may further weaken an already strained homeostatic regulation and thereby significantly increase the risk of serious clinical complications of TLS.5 Elderly patients (age > 65 years) with cancer are particularly likely to have comorbidities, which may worsen their prognosis in the event of TLS, including baseline chronic renal insufficiency and/or heart disease.6
Hyperuricemia is one of the hallmarks of LTLS. If not reversed quickly, severe hyperuricemia may have serious clinical consequences, particularly acute kidney injury (AKI), which is an independent risk factor for mortality (even mild AKI).2 Additionally, hyperuricemia can lead to a variety of distressing symptoms, such as gastrointestinal complaints (nausea, vomiting, diarrhea, and anorexia), lethargy, hematuria, flank or back pain, fluid overload, edema, arthralgias, hypertension, and signs of obstructive uropathy.7
The risk of renal complications is particularly high in elderly patients with baseline renal disease, which may have been caused by diabetes, hypertension, renal artery stenosis, chronic pyelonephritis, amyloidosis, glomerular disease, or treated malignancies.6 Furthermore, congestive heart failure, use of thiazide or loop diuretics, obesity, type II diabetes, renal impairment, hypertriglyceridemia, and peripheral vascular disease are major cardiovascular risk factors associated with hyperuricemia.6
Serious clinical consequences may arise not only from hyperuricemia but also from TLS-related electrolyte abnormalities (ie, hyperphosphatemia, hyperkalemia, or hypocalcemia). AKI, cardiac arrhythmia, and neurologic impairment, such as seizures or higher CNS dysfunction, may result from severe hyperphosphatemia and secondary hypocalcemia caused by calcium phosphate precipitation in renal tubules. These two metabolic disturbances also may present with muscle cramps, tetany, or perioral numbness or tingling; rare symptoms include fatigue, bone and joint pain, pruritus, and rash.7 Hyperphosphatemia can be exacerbated by excessive use of phosphate-containing laxatives or enemas, which is especially common in the elderly. Hyperkalemia also can lead to neuromuscular symptoms, but principally, it may cause potentially life-threatening cardiac dysfunction.2
Because of the decrease in physiologic reserves with age, the aforementioned electrolyte abnormalities are associated with increased morbidity and mortality in the elderly.8 For example, renal function generally declines with age, and age-related reduction in renin and aldosterone levels increases the risk of hyperkalemia.6 Particularly among the elderly, the risk of hyperkalemia is increased in those with renal tubular reabsorption/secretion defects, those with type II diabetes who develop type IV renal tubular acidosis as the result of hyporeninemic hypoaldosteronism, and those who take nonsteroidal anti-inflammatory drugs on a long-term, scheduled basis.9,10 Although antihypertensive medications generally have cardioprotective and renoprotective properties, many antihypertensives actually may increase the risk of hyperkalemia in elderly patients with renal tubular acidosis.11,12 Also in adults, hypocalcemia may result from vitamin D deficiency, impaired vitamin D metabolism, low intestinal Ca2+ absorption, phosphate retention, chronic hypomagnesemia, serum protein abnormalities, and parathyroid hormone resistance; it may also occur as a medication adverse effect (specifically relevant for patients with cancer treated with bisphosphonates, anticonvulsants, cis¬platin, and the combination of 5-fluor¬¬ouracil and ¬leucovorin).6,13
Risk of TLS in adults with cancer
The risk of TLS in adults with cancer depends on multiple components, including disease-related factors, the type and aggressiveness of anticancer treatment, other medications concomitantly administered, and patient (host)-related factors. TLS has been observed primarily in patients with hematologic malignancies, with particularly high incidences reported for those with Burkitt (and Burkitt-like) lymphoma, precursor B-lymphoblastic leukemia/lymphoma, high-stage T-cell anaplastic large cell lymphoma, and ALL.2
Demographic data from compassionate-use studies of the uricolytic agent rasburicase (Elitek) in patients with or at high risk of acute cancer-associated hyperuricemia and TLS seem to suggest that AML with high WBC counts and select types of NHL (mostly high-grade and some intermediate-grade lymphomas) are associated with a high risk of TLS in adults.14,15 Based on the findings of two European studies, between 3% and 17% of adult patients with AML may experience LTLS or CTLS in response to induction therapy.4,16 In one of the two studies, approximately 20% of adults with AML, ALL, or NHL experienced hyperuricemia after induction therapy.4 Data from the second study, which included 772 adults with AML (including some cases of chronic myelogenous leukemia [CML] in blast crisis) who had received induction chemotherapy, were used to develop a risk-prediction model for CTLS in adult patients with AML.16 According to the model, high WBC count, pretreatment hyperuricemia, and high baseline serum creatinine and LDH concentrations were significant independent prognostic factors for the development of LTLS and CTLS.16
An independent international panel of experts in pediatric and adult hematologic malignancies and TLS recently developed guidelines for the management of TLS based on a comprehensive risk-stratification algorithm. These guidelines were published in 2008 by the American Society of Clinical Oncology.17 Patients were considered to be at high, intermediate, or low risk of TLS, depending on the specific type of malignancy, tumor burden, and type of cytoreductive therapy administered.
In addition to these basic risk categories, other risk factors such as renal function and plasma uric acid (PUA) level at baseline were incorporated into the recommendations for TLS prevention and treatment.17 For example, the presence of baseline hyperuricemia (defined as a serum uric acid level > 7.5 mg/dL) is a modifier of the recommendation of antihyperuricemic therapy for patients at intermediate risk of TLS; if there are high baseline PUA levels, the drug of choice should not be allopurinol but rather rasburicase.17 However, these guidelines do not address all malignancies or uniformly assess risk depending on renal involvement by the disease or kidney function.
Consequently, another consensus panel was convened to build upon the 2008 guidelines and produce a medical decision tree for ranking patients with cancer as low, intermediate, or high risk of TLS. For this project, risk factors included biologic evidence of LTLS, tumor proliferation, and bulk and stage of malignant tumor, as well as renal impairment and/or involvement by the disease at the time of TLS diagnosis; subsequently, an algorithmic model of low-, intermediate-, and high-risk TLS classification and associated TLS prophylaxis recommendations were finalized.18 TLS risk factors of particular relevance for elderly patients are age-related alterations in heart anatomy and function, obesity, generalized deconditioning, alterations of the cardiovascular and circulatory systems, use of multiple drugs with potential pharmacodynamic interactions, age-related decrease in glomerular filtration rate, tobacco use, excessive alcohol consumption, and unhealthy dietary ¬habits.6
In addition to hematologic malignancies, solid tumors may result in TLS as a response to chemo(bio)-therapy or radiation/chemoradiation therapy or spontaneously. In adults, solid tumors that have been associated with TLS include, but are not limited to, breast cancer, lung cancer (especially small cell lung cancer), gastrointestinal stromal tumor, germ cell tumor, hepatocellular carcinoma, melanoma, malignant pheochromocytoma, ovarian cancer, colon cancer, and renal cell carcinoma.19–21 Table 1 presents a list of recent reports of TLS in adults with hematologic malignancies or solid tumors.22–41 Although TLS is less common in patients with solid tumors than in those with hematologic malignancies, its onset and progression in patients with solid tumors often are less predictable.19 Consequently, patients with TLS from solid tumors tend to have worse prognoses.19 In patients with solid tumors, TLS may develop days or even weeks after initiation of chemotherapy, providing no or limited opportunity for effective prophylaxis. Thus, for patients with solid tumors, heightened awareness of and vigilance for TLS are necessary to ensure appropriate and timely management of TLS.
The importance of such timely management of TLS is illustrated by several case studies. In one such case, a 55-year-old man with advanced hepatocellular carcinoma developed acute renal failure, hyperkalemia, and hyperuricemia 30 days after the initiation of treatment with the oral tyrosine kinase inhibitor sorafenib (Nexavar). The patient eventually died of multiple organ failure, despite treatment with hyperhydration, administration of the maximal daily dose of allopurinol, and use of emergency hemodialysis.37
Another fatal outcome of TLS was reported for a 62-year-old man with metastatic colon cancer and a 10-year complicated history of treatments. The patient, who had extensive lung metastases, hyperuricemia, and elevated serum LDH and creatinine concentrations at baseline, developed severe TLS within 2 days of receiving bevacizumab (Avastin) with combination chemotherapy and eventually died of acute renal failure.34
Furthermore, a rare case of spontaneous TLS occurred in a patient with Crohn’s disease who developed plasmacytoma while being treated with immunosuppressants. The patient experienced extreme hyperuricemia (a PUA level of 44 mg/dL) and died of the consequences of acute oliguric renal failure, despite hyperhydration, alkalinization, treatment with maximal doses of allopurinol followed by rasburicase, and hemodialysis.39
According to current TLS management guidelines, patients with multiple myeloma or rapidly growing solid tumors and an expected rapid response to therapy are considered to be at intermediate risk of TLS. However, the risk of TLS in patients with solid tumors is increased by the presence of bulky disease (masses or lymph nodes > 10 cm),17 unfavorable host baseline characteristics (such as renal insufficiency and baseline hyperuricemia),17 and the presence of liver metastases.19 According to relatively recent case reports, TLS occurred in two adult patients with hepatocellular carcinoma undergoing transarterial chemoembolization36 and in a 58-year-old man with metastatic melanoma and bulky liver metastases, who developed TLS within 24 hours after receiving intra-arterial infusion of cisplatin and embolization therapy.42 TLS from palliative radiotherapy, although rare, also has been reported; two fatal cases include a 74-year-old man with diffuse large B-cell lymphoma43 and a 52-year-old man with non-small cell lung cancer.38
Chemo(bio)therapy regimens associated with risk of TLS
The availability of new agents (either as monotherapy or in combination regimens) with high tumor response rates is expected to increase the risk and incidence of TLS. Even for hematologic malignancies with a relatively low incidence of TLS, such as chronic lymphocytic leukemia (CLL), the risk of TLS may increase with the growing use of novel agents that are highly effective inducers of apoptosis.44
In CLL associated with WBC counts of 10,000–100,000 cells/µL, fludarabine therapy (as monotherapy or combination therapy) confers intermediate-risk status (for TLS), according to current TLS management guidelines.17 Current US prescribing information for rituximab (Rituxan), which together with fludara¬bine (± cyclophosphamide) constitutes the standard treatment backbone for previously untreated CLL,45 contains a black-box warning for TLS when rituximab is given for the treatment of NHL.46 A warning for TLS also has been issued for the use of bendamustine (Treanda) for CLL.47 Alvocidib (flavopiridol), a cyclin-dependent kinase inhibitor in clinical development (phase II studies) for the treatment of adults with relapsed CLL, has been associated with a high incidence of TLS, particularly in patients with WBC counts > 200,000 cells/µL.48,49 Finally, lenalidomide (Revlimid) was associated with TLS and a tumor flare phenomenon in patients with CLL in phase II clinical studies.50
A number of case reports have associated imatinib (Gleevec), bortezomib (Velcade), and thalidomide (Thalomid) with the development of TLS in adults. Administration of imatinib led to TLS in two patients treated for ALL and in one patient treated for CML.23 Bortezomib and/or thalidomide caused TLS in several patients treated for multiple myeloma.31,51–54 TLS also is common in patients receiving chemotherapy for acute adult human T-cell lymphotrophic virus-1–associated T-cell leukemia/lymphoma (ATLL). Fatal TLS recently occurred in an obese 57-year-old woman with ATLL, in the background of systemic lupus erythematosus, after chemotherapy with high-dose prednisone and adjusted doses of cyclophosphamide and doxorubicin.33
Management of hyperuricemia in adults: the role of rasburicase
Because hyperuricemia can increase both the risk and the severity of CTLS, management of hyperuricemia that focuses on its prevention and, in cases where it has already emerged, its rapid reversal is an important strategy in reducing TLS-associated mortality and morbidity. The current recommendations for the prevention and management of TLS-associated hyperuricemia have been reviewed in great detail recently.2 Monitoring and clinical judgment generally are adequate for low-risk patients, but patients with higher risk require hydration and antihyperuricemic management with appropriate drug therapy.2 For most patients with an intermediate risk of TLS, prophylactic antihyperuricemic therapy with allopurinol is sufficient, because allopurinol can prevent the buildup of uric acid by blocking the enzymatic conversion of hypoxanthine and xanthine to uric acid (via direct inhibition of xanthine oxidase). However, allopurinol does not modify existing uric acid pools and thus is inadequate for the prevention or management of hyperuricemia in high-risk patients.2 Because a TLS-associated rise in uric acid levels can occur suddenly and rapidly in patients at high risk, effective and safe degradation of uric acid is essential for preventing or reversing life-threatening hyperuricemia.
The effectiveness of rasburicase as antihyperuricemic therapy for adult patients with cancer was demonstrated initially in two international multicenter compassionate-use studies. One study, conducted in the United States and Canada, included 387 adults (and 682 children) with mostly hematologic malignancies who received daily 30-minute IV administrations of rasburicase for 1–7 days at a dose of 0.2 mg/kg.15 All patients who received rasburicase for TLS prophylaxis, including 126 adults, maintained low PUA levels during chemotherapy, and all 212 adults with hyperuricemia at baseline responded to treatment with prompt and impressive reduction in PUA levels, which was maintained for several days.15 Rasburicase generally was well tolerated; only two adults experienced a single episode each of grade 3 toxicity, and no adult experienced grade 4 toxicity.15
The second study—a multicenter compassionate-use study in patients with leukemia or lymphoma from Europe and Australia—included 112 adults (and 166 children) with generally large tumor burdens, as assessed by WBC counts for leukemia and clinical stage and also by serum LDH levels for lymphoma.14 Reduction in PUA levels with rasburicase was statistically and clinically highly significant, resulting in PUA response rates of 100% in both adults and children. Only one adult patient, a 41-year-old man with AML and hyperuricemia at baseline, required hemodialysis because of acute renal failure, although PUA levels returned to normal after treatment with rasburicase. The patient subsequently died of respiratory failure attributable to disease ¬progression.14
The multicenter US registrational trial for the adult indication of rasburicase included 280 patients with hematologic malignancies who were at risk of TLS.55 Patients were randomly assigned to one of three treatments: rasburicase (0.2 mg/kg/d given as a 30-minute IV infusion) for 5 days, rasburicase for 3 days followed by oral allopurinol (300 mg/d) for 2 days, or allopurinol for 5 days. Rasburicase alone achieved significantly greater response rates than allopurinol monotherapy both in patients with a high risk of TLS and in those with baseline hyperuricemia (Table 2). In this clinical trial, control of PUA concentrations was achieved within 4 hours with rasburicase (compared with a median time interval of 27 hours with allopurinol) and was maintained during the 7-day monitoring period after initiation of antihyperuricemic therapy. Although reduction in the incidence of TLS was not an efficacy endpoint of the study, safety data analysis revealed that the occurrence of LTLS or CTLS was less common in the rasburicase than in the allopurinol treatment arm.55
In this large, randomized clinical study, rasburicase was safe and generally well tolerated during the 5-day treatment period. Patients who received rasburicase and those who received allopurinol had a similar incidence of adverse events.55,56 The only drug-related adverse events observed among the 184 patients who received rasburicase (alone or in combination with allopurinol) were potential hypersensitivity reactions in five patients, including irritation at the injection site (one patient); arthralgia, myalgia, and rash (one patient); peripheral edema (one patient); and grade 3 hypersensitivity (two patients). One patient with grade 3 hypersensitivity related to rasburicase discontinued study participation on day 1. No grade 4 hypersensitivity reaction, anaphylaxis, hemolytic reaction, or methemoglobinemia occurred with rasburicase.55
In summary, the results of this phase III randomized controlled study show that rasburicase may be superior to allopurinol in preventing hyperuricemia in adult leukemia/lymphoma patients at high risk of TLS and in achieving rapid and effective control of PUA levels in patients with hyperuricemia. The results support the recommendation within the current TLS management guidelines of using rasburicase for prophylaxis in high-risk patients and for treatment of hyperuricemia of malignancy, as well as in those patients with fully developed LTLS or CTLS (irrespective of PUA levels).17
Conclusion
In adult patients with cancer, the risk of serious clinical consequences of TLS often is increased by the presence of renal impairment, heart disease, baseline metabolic imbalances, and multiorgan derangements. Even in adult patients without serious comorbidities, an age-related decrease in physiologic resistance (reduced pathobiologic reserve), lifestyle factors (eg, adverse chronic exposure to tobacco, alcohol, or other toxins; sedentary existence; obesity; and vitamin and macro/micronutrient deficiencies), and dependence on multiple medications (polypharmacy and an associated increased risk for drug-drug interactions) should be considered for both TLS risk assessment and therapeutic choices in TLS prophylaxis and treatment. Although TLS is most common in patients with hematologic malignancies, serious cases of TLS have been reported in patients with a large variety of solid tumors, with sometimes fatal outcomes.
Assessment of TLS risk in patients with solid tumors, who generally tend to belong to a more aged population, is more difficult than in those with hematologic malignancies. Tumor burden and the type of chemotherapy are the most important risk factors for development of TLS in association with solid tumors; however, an unexpected occurrence of acute spontaneous TLS with hyperuricemia has been observed, even in the absence of these two factors.
Hyperuricemia is a key manifestation of TLS and is associated with risks of AKI, fluid overload, heart failure, and death. Prevention of hyperuricemia in high-risk patients and its rapid reversal in those with hyperuricemia at presentation or in those with fully developed TLS are of utmost importance in the successful management of TLS in adults with cancer. Results of the recent phase III study of rasburicase in adult patients with cancer demonstrated that rasburicase is superior to allopurinol in providing rapid and effective control of PUA levels.
Acknowledgments: Editorial assistance provided by Roland Tacke, PhD, and Candace Lundin, DVM, MS, was funded by sanofi-aventis US. The authors were fully responsible for all content and editorial decisions and did not receive financial support or compensation related to the development of this article.
References
- Cairo MS, Bishop M. Tumour lysis syndrome: new therapeutic strategies and classification. Br J Haematol 2004;127:3–11.
- Mughal TI, Ejaz AA, Foringer JR, Coiffier B. An integrated clinical approach for the identification, prevention, and treatment of tumor lysis syndrome. Cancer Treat Rev 2010;36:164–176.
- Hande KR, Garrow GC. Acute tumor lysis syndrome in patients with high-grade non-Hodgkin’s lymphoma. Am J Med 1993;94:133–139.
- Annemans L, Moeremans K, Lamotte M, et al. Incidence, medical resource utilisation and costs of hyperuricemia and tumour lysis syndrome in patients with acute leukaemia and non-Hodgkin’s lymphoma in four European countries. Leuk Lymphoma 2003;44:77–83.
- Coiffier B, Riouffol C. Management of tumor lysis syndrome in adults. Expert Rev Anticancer Ther 2007;7:233–239.
- Pumo V, Sciacca D, Malaquarnera M. Tumor lysis syndrome in elderly. Crit Rev ¬Oncol Hematol 2007;64:31–42.
- Kennedy LD, Ajiboye VO. Rasburicase for the prevention and treatment of hyperuricemia in tumor lysis syndrome. J Oncol Pharm Pract 2010;16:205–213.
- Allison SP, Lobo DN. Fluid and electrolytes in the elderly. Curr Opin Clin Nutr Metab Care 2004;7:27–33.
- Michelis MF. Hyperkalemia in the elderly. Am J Kidney Dis 1990;16:296–299.
- Williams ME. Endocrine crises: hyperkalemia. Crit Care Clin 1991;7:155–174.
- Weir MR. Non-diuretic-based antihypertensive therapy and potassium homeostasis in elderly patients. Coron Artery Dis 1997;8:499–504.
- Sica DA. Antihypertensive therapy and its effects on potassium homeostasis. J Clin Hypertens (Greenwich) 2006;8:67–73.
- Moe SM. Disorders involving calcium, phosphorus, and magnesium. Prim Care 2008;35:215–237.
- Bosly A, Sonet A, Pinkerton CR, et al. Rasburicase (recombinant urate oxidase) for the management of hyperuricemia in patients with cancer: report of an international compassionate use study. Cancer 2003;98:1048–1054.
- Jeha S, Kantarjian H, Irwin D, et al. Efficacy and safety of rasburicase, a recombinant urate oxidase (Elitek), in the management of malignancy-associated hyperuricemia in pediatric and adult patients: final results of a multicenter compassionate use trial. Leukemia 2005;19:34–38.
- Montesinos P, Lorenzo I, Martin G, et al. Tumor lysis syndrome in patients with acute myeloid leukemia: identification of risk factors and development of a predictive model. Haematologica 2008;93:67–74.
- Coiffier B, Altman A, Pui CH, Younes A, Cairo MS. Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review. J Clin Oncol 2008;26:2767–2778.
- Cairo MS, Coiffier B, Reiter A, Younes A; TLS Expert Panel. Recommendations for the evaluation of risk and prophylaxis of tumour lysis syndrome (TLS) in adults and children with malignant diseases: an expert TLS panel consensus. Br J Haematol 2010;149:578–586.
- Gemici C. Tumour lysis syndrome in solid tumours. Clin Oncol (R Coll Radiol) 2006;18:773–780.
- Pinder EM, Atwal GS, Ayantunde AA, et al. Tumour lysis syndrome occurring in a patient with metastatic gastrointestinal stromal tumour treated with Glivec (imatinib mesylate, Gleevec, STI571). Sarcoma 2007;2007:82012.
- Mott FE, Esana A, Chakmakjian C, Herrington JD. Tumor lysis syndrome in solid tumors. Support Cancer Ther 2005;2:188–191.
- Konuma T, Ooi J, Takahashi S, et al. Fatal acute tumor lysis syndrome following intrathecal chemotherapy for acute lymphoblastic leukemia with meningeal involvement. Intern Med 2008;47:1987–1988.
- Chang H, Shih LY. Imatinib-induced tumor lysis syndrome: report of a case and review of the literature. Chang Gung Med J 2008;31:510–514.
- Rüger W, Kruip MJ, Betjes MG. Reversible renal failure due to bilateral renal sarcoma in a patient with acute myeloid leukemia. Ren Fail 2009;31:606–609.
- Kosseifi SG, Vyas B, Vyas H, Krishnaswamy G, Smith S. Tumor lysis syndrome following chlorambucil therapy for chronic lymphocytic leukemia: case report and review of the literature. Tenn Med 2009;102:33–36.
- Calvo-Villas JM, Urcuyo BM, Umpierrez AM, Sicilia F. Acute tumor lysis syndrome during oral fludarabine treatment for chronic lymphocytic leukemia: role of treatment with rasburicase. Onkologie 2008;31:197–199.
- Al-Kali A, Farooq S, Tfayli A. Tumor lysis syndrome after starting treatment with Gleevec in a patient with chronic myelogenous leukemia. J Clin Pharm Ther 2009;34:607–610.
- Sonmez M, Ovali E, Omay SB. Tumor lysis syndrome during treatment with AMN107 (nilotinib) in a patient with chronic myelogenous leukemia accelerated phase. J Clin Pharm Ther 2008;33:91–92.
- Lin CJ, Chen HH, Hsieh RK, Chen YC, Wu CJ. Acute tumor lysis syndrome in a hemodialysis patient with diffuse large B cell lymphoma. Med Oncol 2009;26:93–95.
- Kasahara I, Nishio M, Yamamoto S, et al. Cord blood transplantation with a reduced-intensity conditioning regimen for patients with relapsed aggressive multiple myeloma after cytoreduction with bortezomib. Int J Hematol 2009;90:413–415.
- Chim CS. Rapid complete remission in multiple myeloma with bortezomib/thalidomide/dexamethasone combination therapy following development of tumor lysis syndrome. Cancer Chemother Pharmacol 2008;62:181–182.
- Bouaziz JD, Cordel N, Hickman G, Fieschi C, Ortonne N, Bagot M. Cutaneous tumor lysis syndrome in a patient with HTLV-1 adult T-cell lymphoma/leukemia. Blood 2009;114:4320–4321.
- Fritsch-Stork RD, Leguit RJ, Derksen RH. Rapidly fatal HTLV-1-associated T-cell leukemia/lymphoma in a patient with SLE. Nat Rev Rheumatol 2009;5:283–287.
- Hentrich M, Schiel X, Scheidt B, Reitmeier M, Hoffmann U, Lutz L. Fatal tumor lysis syndrome after irinotecan/5-FU/folinic acid/bevacizumab-containing therapy in a patient heavily pretreated for metastatic colon cancer. Acta Oncol 2008;47:155–156.
- Feres GA, Salluh JI, Ferreira CG, Soares M. Severe acute tumor lysis syndrome in patients with germ-cell tumors. Indian J Urol 2008;24:555–557.
- Hsieh PM, Hung KC, Chen YS. Tumor lysis syndrome after transarterial chemoembolization of hepatocellular carcinoma: case reports and literature review. World J Gastroenterol 2009;15:4726–4728.
- Huang WS, Yang CH. Sorafenib induced tumor lysis syndrome in an advanced hepatocellular carcinoma patient. World J Gastroenterol 2009;15:4464–4466.
- Noh GY, Choe DH, Kim CH, Lee JC. Fatal tumor lysis syndrome during radiotherapy for non-small-cell lung cancer. J Clin Oncol 2008;26:6005–6006.
- Froilán Torres C, Castro Carbajo P, Pajares Villarroya R, et al. Acute spontaneous tumor lysis syndrome in a patient with Crohn’s disease taking immunosuppressants. Rev Esp Enferm Dig 2009;101:288–294.
- Michels J, Lassau N, Gross-Goupil M, Massard C, Mejean A, Escudier B. Sunitinib inducing tumor lysis syndrome in a patient treated for renal carcinoma. Invest New Drugs 2010;28:690–693.
- Shenoy C. Acute spontaneous tumor lysis syndrome in a patient with squamous cell carcinoma of the lung. QJM 2009;102:71–73.
- Nakamura Y, Nakamura Y, Hori E, et al. Tumor lysis syndrome after transcatheter arterial infusion of cisplatin and embolization therapy for liver metastases of melanoma. Int J Dermatol 2009;48:763–767.
- Yamazaki H, Hanada M, Horiki M, et al. Acute tumor lysis syndrome caused by palliative radiotherapy in patients with diffuse large B-cell lymphoma. Radiat Med 2004;22:52–55.
- Cheson BD. Etiology and management of tumor lysis syndrome in patients with chronic lymphocytic leukemia. Clin Adv Hematol Oncol 2009;7:263–271.
- Lin TS. Novel agents in chronic lymphocytic leukemia: efficacy and tolerability of new therapies. Clin Lymphoma Myeloma 2008;8(suppl 4):S137–S143.
- Rituxan [package insert]. South San Francisco, CA: Genentech, Inc.; 2011.
- Treanda [package insert]. Frazer, PA: Cephalon, Inc.; 2010.
- Lin TS, Ruppert AS, Johnson AJ, et al. Phase II study of flavopiridol in relapsed chronic lymphocytic leukemia demonstrating high response rates in genetically high-risk disease. J Clin Oncol 2009;27:6012–6018.
- Byrd JC, Lin TS, Dalton JT, et al. Flavopiridol administered using a pharmacologically derived schedule is associated with marked clinical efficacy in refractory, genetically high-risk chronic lymphocytic leukemia. Blood 2007;109:399–404.
- Moutouh-de Parseval LA, Weiss L, DeLap RJ, Knight RD, Zeldis JB. Tumor lysis syndrome/tumor flare reaction in lenalidomide-treated chronic lymphocytic leukemia. J Clin Oncol 2007;25:5047.
- Sezer O, Vesole DH, Singhal S, et al. Bortezomib-induced tumor lysis syndrome in multiple myeloma. Clin Lymphoma Myeloma 2006;7:233–235.
- Huston A, Brown J, Roodman GD. Tumor lysis syndrome following thalidomide and dexamethasone therapy for newly diagnosed multiple myeloma. Exp Hematol 2006;34:1616.
- Berenson JR, Yang HH, Vescio RA, et al. Safety and efficacy of bortezomib and melphalan combination in patients with relapsed or refractory multiple myeloma: updated results of a phase 1/2 study after longer follow-up. Ann Hematol 2008;87:623–631.
- Fuente N, Mañe JM, Barcelo R, Muñoz A, Perez-Hoyos T, Lopez-Vivanco G. Tumor lysis syndrome in a multiple myeloma patient treated with thalidomide. Ann Oncol 2004;15:537.
- Cortes J, Moore JO, Maziarz RT, et al. Control of plasma uric acid in adults at risk for tumor lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone: results of a multicenter phase III study. J Clin Oncol 2010;28:4207–4213.
- Elitek [package insert]. Bridgewater, NJ: sanofi-aventis U.S., LLC; 2009.
ABOUT THE AUTHORS
Affiliations: Dr. Seiter is Professor of Medicine and Director of the Leukemia Service at New York Medical College, Valhalla, NY. Dr. Sarlis is currently Vice President and Head of the Medical Affairs Department at Incyte Corporation, Wilmington, DE. Dr. Kim is Associate Professor and Chief, Section of Head and Neck Medical Oncology, Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, at The University of Texas MD Anderson Cancer Center, Houston, TX.
Conflicts of interest: Dr. Sarlis was an employee of sanofi-aventis U.S. at the time this article was written and holds stock options and stock in this company. Drs. Seiter and Kim have no pertinent conflicts of interest to disclose.
Karen P. Seiter, MD,1 Nicholas J. Sarlis, MD, PhD, FACP,2* and Edward S. Kim, MD3
- New York Medical College, Valhalla, NY;
- Medical Affairs–Oncology, sanofi-aventis U.S., Bridgewater, NJ; and
- The University of Texas MD Anderson Cancer Center, Houston, TX
Manuscript received September 9, 2010; accepted April 8, 2011.
* Current affiliation: Incyte Corporation, Wilmington, DE.
Correspondence to: Karen P. Seiter, MD, New York Medical College, Munger Pavilion, Room 250, Valhalla, NY 10595; telephone: 914-493-7514; fax: 914-594-4420; e-mail: [email protected].
Tumor lysis syndrome (TLS) is a relatively common, potentially life-threatening complication of aggressive cytotoxic therapy characterized by metabolite and electrolyte abnormalities (eg, hyperuricemia). To increase the awareness of the risk of hyperuricemia and TLS in adult patients with cancer, who are likely to have age- or lifestyle-related comorbidities, the authors examine the pathophysiology and risk of TLS in adult patients with a broad spectrum of cancer diagnoses. Current recommendations for effective prophylaxis and management of TLS are summarized briefly. Particular emphasis is given to the appropriate role of antihyperuricemic therapy with rasburicase in adults, based on the recent results of a phase III clinical study.
Tumor lysis syndrome (TLS) is a serious, potentially life-threatening condition of metabolic derangement and impaired electrolyte homeostasis. TLS can occur in patients with cancer as a result of spontaneous or, more commonly, treatment-induced tumor cell death and typically is seen in patients with hematologic, rather than solid organ, malignancies who are undergoing chemotherapy. It is particularly an issue for patients with rapidly growing tumors and a high tumor burden (as evidenced by a high white blood cell [WBC] count in leukemia and elevated serum lactate dehydrogenase [LDH] levels and/or advanced clinical stage in lymphoma) at the beginning of chemotherapy. TLS is caused by a sudden, massive release of cell content from lysed tumor cells into the bloodstream, which overwhelms the body’s capacity for homeostatic regulation. Because persistent or progressive metabolic derangement is associated with a high risk of organ failure, TLS is a clinical emergency.
To facilitate the diagnosis, prevention, and treatment of TLS, Cairo and Bishop established precise criteria for the categorization of TLS on the basis of metabolic abnormalities and the associated significant clinical toxicities that require clinical intervention.1 According to Cairo and Bishop, laboratory TLS (LTLS) is defined by the presence of two or more of the following metabolic abnormalities: hyperuricemia, hyperkalemia, hyperphosphatemia, and secondary hypocalcemia. Clinical TLS (CTLS) is defined as LTLS accompanied by at least one clinical complication, such as renal impairment (defined as a serum creatinine concentration greater than 1.5 times the upper limit of normal), cardiac arrhythmia, or seizure. The aforementioned clinical manifestations should not be directly or likely attributable to a therapeutic agent (eg, a rise in creatinine levels after administration of a nephrotoxic drug).1
Estimates from clinical studies of the incidence of TLS vary widely.2 In a review of case records of 102 patients with high-grade non-Hodgkin’s lymphoma (NHL), 42% of patients had signs of LTLS and 6% had CTLS.3 In contrast, analysis of data from a sample of 755 European children and adults with newly diagnosed or recurrent acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), or NHL indicated a 19% incidence of hyperuricemia and a 5% incidence of LTLS per episode of administration of an induction therapy ¬regimen.4
Pathophysiology of TLS in adults
The pathophysiology and clinical consequences of TLS have been discussed infrequently in the context of adult patient populations. Yet adult ¬patients are more likely than pediatric patients to experience potentially serious metabolic, cardiac, renal, or multisystemic comorbidities. Such comorbidities, whether chronic illnesses present at the initiation of anticancer therapy or acute conditions that develop during administration of aggressive cytoreductive regimens, require special consideration because they may amplify the metabolic and electrolyte imbalances caused by tumor cell lysis and may have secondary pathophysiologic consequences. In adults, these effects may further weaken an already strained homeostatic regulation and thereby significantly increase the risk of serious clinical complications of TLS.5 Elderly patients (age > 65 years) with cancer are particularly likely to have comorbidities, which may worsen their prognosis in the event of TLS, including baseline chronic renal insufficiency and/or heart disease.6
Hyperuricemia is one of the hallmarks of LTLS. If not reversed quickly, severe hyperuricemia may have serious clinical consequences, particularly acute kidney injury (AKI), which is an independent risk factor for mortality (even mild AKI).2 Additionally, hyperuricemia can lead to a variety of distressing symptoms, such as gastrointestinal complaints (nausea, vomiting, diarrhea, and anorexia), lethargy, hematuria, flank or back pain, fluid overload, edema, arthralgias, hypertension, and signs of obstructive uropathy.7
The risk of renal complications is particularly high in elderly patients with baseline renal disease, which may have been caused by diabetes, hypertension, renal artery stenosis, chronic pyelonephritis, amyloidosis, glomerular disease, or treated malignancies.6 Furthermore, congestive heart failure, use of thiazide or loop diuretics, obesity, type II diabetes, renal impairment, hypertriglyceridemia, and peripheral vascular disease are major cardiovascular risk factors associated with hyperuricemia.6
Serious clinical consequences may arise not only from hyperuricemia but also from TLS-related electrolyte abnormalities (ie, hyperphosphatemia, hyperkalemia, or hypocalcemia). AKI, cardiac arrhythmia, and neurologic impairment, such as seizures or higher CNS dysfunction, may result from severe hyperphosphatemia and secondary hypocalcemia caused by calcium phosphate precipitation in renal tubules. These two metabolic disturbances also may present with muscle cramps, tetany, or perioral numbness or tingling; rare symptoms include fatigue, bone and joint pain, pruritus, and rash.7 Hyperphosphatemia can be exacerbated by excessive use of phosphate-containing laxatives or enemas, which is especially common in the elderly. Hyperkalemia also can lead to neuromuscular symptoms, but principally, it may cause potentially life-threatening cardiac dysfunction.2
Because of the decrease in physiologic reserves with age, the aforementioned electrolyte abnormalities are associated with increased morbidity and mortality in the elderly.8 For example, renal function generally declines with age, and age-related reduction in renin and aldosterone levels increases the risk of hyperkalemia.6 Particularly among the elderly, the risk of hyperkalemia is increased in those with renal tubular reabsorption/secretion defects, those with type II diabetes who develop type IV renal tubular acidosis as the result of hyporeninemic hypoaldosteronism, and those who take nonsteroidal anti-inflammatory drugs on a long-term, scheduled basis.9,10 Although antihypertensive medications generally have cardioprotective and renoprotective properties, many antihypertensives actually may increase the risk of hyperkalemia in elderly patients with renal tubular acidosis.11,12 Also in adults, hypocalcemia may result from vitamin D deficiency, impaired vitamin D metabolism, low intestinal Ca2+ absorption, phosphate retention, chronic hypomagnesemia, serum protein abnormalities, and parathyroid hormone resistance; it may also occur as a medication adverse effect (specifically relevant for patients with cancer treated with bisphosphonates, anticonvulsants, cis¬platin, and the combination of 5-fluor¬¬ouracil and ¬leucovorin).6,13
Risk of TLS in adults with cancer
The risk of TLS in adults with cancer depends on multiple components, including disease-related factors, the type and aggressiveness of anticancer treatment, other medications concomitantly administered, and patient (host)-related factors. TLS has been observed primarily in patients with hematologic malignancies, with particularly high incidences reported for those with Burkitt (and Burkitt-like) lymphoma, precursor B-lymphoblastic leukemia/lymphoma, high-stage T-cell anaplastic large cell lymphoma, and ALL.2
Demographic data from compassionate-use studies of the uricolytic agent rasburicase (Elitek) in patients with or at high risk of acute cancer-associated hyperuricemia and TLS seem to suggest that AML with high WBC counts and select types of NHL (mostly high-grade and some intermediate-grade lymphomas) are associated with a high risk of TLS in adults.14,15 Based on the findings of two European studies, between 3% and 17% of adult patients with AML may experience LTLS or CTLS in response to induction therapy.4,16 In one of the two studies, approximately 20% of adults with AML, ALL, or NHL experienced hyperuricemia after induction therapy.4 Data from the second study, which included 772 adults with AML (including some cases of chronic myelogenous leukemia [CML] in blast crisis) who had received induction chemotherapy, were used to develop a risk-prediction model for CTLS in adult patients with AML.16 According to the model, high WBC count, pretreatment hyperuricemia, and high baseline serum creatinine and LDH concentrations were significant independent prognostic factors for the development of LTLS and CTLS.16
An independent international panel of experts in pediatric and adult hematologic malignancies and TLS recently developed guidelines for the management of TLS based on a comprehensive risk-stratification algorithm. These guidelines were published in 2008 by the American Society of Clinical Oncology.17 Patients were considered to be at high, intermediate, or low risk of TLS, depending on the specific type of malignancy, tumor burden, and type of cytoreductive therapy administered.
In addition to these basic risk categories, other risk factors such as renal function and plasma uric acid (PUA) level at baseline were incorporated into the recommendations for TLS prevention and treatment.17 For example, the presence of baseline hyperuricemia (defined as a serum uric acid level > 7.5 mg/dL) is a modifier of the recommendation of antihyperuricemic therapy for patients at intermediate risk of TLS; if there are high baseline PUA levels, the drug of choice should not be allopurinol but rather rasburicase.17 However, these guidelines do not address all malignancies or uniformly assess risk depending on renal involvement by the disease or kidney function.
Consequently, another consensus panel was convened to build upon the 2008 guidelines and produce a medical decision tree for ranking patients with cancer as low, intermediate, or high risk of TLS. For this project, risk factors included biologic evidence of LTLS, tumor proliferation, and bulk and stage of malignant tumor, as well as renal impairment and/or involvement by the disease at the time of TLS diagnosis; subsequently, an algorithmic model of low-, intermediate-, and high-risk TLS classification and associated TLS prophylaxis recommendations were finalized.18 TLS risk factors of particular relevance for elderly patients are age-related alterations in heart anatomy and function, obesity, generalized deconditioning, alterations of the cardiovascular and circulatory systems, use of multiple drugs with potential pharmacodynamic interactions, age-related decrease in glomerular filtration rate, tobacco use, excessive alcohol consumption, and unhealthy dietary ¬habits.6
In addition to hematologic malignancies, solid tumors may result in TLS as a response to chemo(bio)-therapy or radiation/chemoradiation therapy or spontaneously. In adults, solid tumors that have been associated with TLS include, but are not limited to, breast cancer, lung cancer (especially small cell lung cancer), gastrointestinal stromal tumor, germ cell tumor, hepatocellular carcinoma, melanoma, malignant pheochromocytoma, ovarian cancer, colon cancer, and renal cell carcinoma.19–21 Table 1 presents a list of recent reports of TLS in adults with hematologic malignancies or solid tumors.22–41 Although TLS is less common in patients with solid tumors than in those with hematologic malignancies, its onset and progression in patients with solid tumors often are less predictable.19 Consequently, patients with TLS from solid tumors tend to have worse prognoses.19 In patients with solid tumors, TLS may develop days or even weeks after initiation of chemotherapy, providing no or limited opportunity for effective prophylaxis. Thus, for patients with solid tumors, heightened awareness of and vigilance for TLS are necessary to ensure appropriate and timely management of TLS.
The importance of such timely management of TLS is illustrated by several case studies. In one such case, a 55-year-old man with advanced hepatocellular carcinoma developed acute renal failure, hyperkalemia, and hyperuricemia 30 days after the initiation of treatment with the oral tyrosine kinase inhibitor sorafenib (Nexavar). The patient eventually died of multiple organ failure, despite treatment with hyperhydration, administration of the maximal daily dose of allopurinol, and use of emergency hemodialysis.37
Another fatal outcome of TLS was reported for a 62-year-old man with metastatic colon cancer and a 10-year complicated history of treatments. The patient, who had extensive lung metastases, hyperuricemia, and elevated serum LDH and creatinine concentrations at baseline, developed severe TLS within 2 days of receiving bevacizumab (Avastin) with combination chemotherapy and eventually died of acute renal failure.34
Furthermore, a rare case of spontaneous TLS occurred in a patient with Crohn’s disease who developed plasmacytoma while being treated with immunosuppressants. The patient experienced extreme hyperuricemia (a PUA level of 44 mg/dL) and died of the consequences of acute oliguric renal failure, despite hyperhydration, alkalinization, treatment with maximal doses of allopurinol followed by rasburicase, and hemodialysis.39
According to current TLS management guidelines, patients with multiple myeloma or rapidly growing solid tumors and an expected rapid response to therapy are considered to be at intermediate risk of TLS. However, the risk of TLS in patients with solid tumors is increased by the presence of bulky disease (masses or lymph nodes > 10 cm),17 unfavorable host baseline characteristics (such as renal insufficiency and baseline hyperuricemia),17 and the presence of liver metastases.19 According to relatively recent case reports, TLS occurred in two adult patients with hepatocellular carcinoma undergoing transarterial chemoembolization36 and in a 58-year-old man with metastatic melanoma and bulky liver metastases, who developed TLS within 24 hours after receiving intra-arterial infusion of cisplatin and embolization therapy.42 TLS from palliative radiotherapy, although rare, also has been reported; two fatal cases include a 74-year-old man with diffuse large B-cell lymphoma43 and a 52-year-old man with non-small cell lung cancer.38
Chemo(bio)therapy regimens associated with risk of TLS
The availability of new agents (either as monotherapy or in combination regimens) with high tumor response rates is expected to increase the risk and incidence of TLS. Even for hematologic malignancies with a relatively low incidence of TLS, such as chronic lymphocytic leukemia (CLL), the risk of TLS may increase with the growing use of novel agents that are highly effective inducers of apoptosis.44
In CLL associated with WBC counts of 10,000–100,000 cells/µL, fludarabine therapy (as monotherapy or combination therapy) confers intermediate-risk status (for TLS), according to current TLS management guidelines.17 Current US prescribing information for rituximab (Rituxan), which together with fludara¬bine (± cyclophosphamide) constitutes the standard treatment backbone for previously untreated CLL,45 contains a black-box warning for TLS when rituximab is given for the treatment of NHL.46 A warning for TLS also has been issued for the use of bendamustine (Treanda) for CLL.47 Alvocidib (flavopiridol), a cyclin-dependent kinase inhibitor in clinical development (phase II studies) for the treatment of adults with relapsed CLL, has been associated with a high incidence of TLS, particularly in patients with WBC counts > 200,000 cells/µL.48,49 Finally, lenalidomide (Revlimid) was associated with TLS and a tumor flare phenomenon in patients with CLL in phase II clinical studies.50
A number of case reports have associated imatinib (Gleevec), bortezomib (Velcade), and thalidomide (Thalomid) with the development of TLS in adults. Administration of imatinib led to TLS in two patients treated for ALL and in one patient treated for CML.23 Bortezomib and/or thalidomide caused TLS in several patients treated for multiple myeloma.31,51–54 TLS also is common in patients receiving chemotherapy for acute adult human T-cell lymphotrophic virus-1–associated T-cell leukemia/lymphoma (ATLL). Fatal TLS recently occurred in an obese 57-year-old woman with ATLL, in the background of systemic lupus erythematosus, after chemotherapy with high-dose prednisone and adjusted doses of cyclophosphamide and doxorubicin.33
Management of hyperuricemia in adults: the role of rasburicase
Because hyperuricemia can increase both the risk and the severity of CTLS, management of hyperuricemia that focuses on its prevention and, in cases where it has already emerged, its rapid reversal is an important strategy in reducing TLS-associated mortality and morbidity. The current recommendations for the prevention and management of TLS-associated hyperuricemia have been reviewed in great detail recently.2 Monitoring and clinical judgment generally are adequate for low-risk patients, but patients with higher risk require hydration and antihyperuricemic management with appropriate drug therapy.2 For most patients with an intermediate risk of TLS, prophylactic antihyperuricemic therapy with allopurinol is sufficient, because allopurinol can prevent the buildup of uric acid by blocking the enzymatic conversion of hypoxanthine and xanthine to uric acid (via direct inhibition of xanthine oxidase). However, allopurinol does not modify existing uric acid pools and thus is inadequate for the prevention or management of hyperuricemia in high-risk patients.2 Because a TLS-associated rise in uric acid levels can occur suddenly and rapidly in patients at high risk, effective and safe degradation of uric acid is essential for preventing or reversing life-threatening hyperuricemia.
The effectiveness of rasburicase as antihyperuricemic therapy for adult patients with cancer was demonstrated initially in two international multicenter compassionate-use studies. One study, conducted in the United States and Canada, included 387 adults (and 682 children) with mostly hematologic malignancies who received daily 30-minute IV administrations of rasburicase for 1–7 days at a dose of 0.2 mg/kg.15 All patients who received rasburicase for TLS prophylaxis, including 126 adults, maintained low PUA levels during chemotherapy, and all 212 adults with hyperuricemia at baseline responded to treatment with prompt and impressive reduction in PUA levels, which was maintained for several days.15 Rasburicase generally was well tolerated; only two adults experienced a single episode each of grade 3 toxicity, and no adult experienced grade 4 toxicity.15
The second study—a multicenter compassionate-use study in patients with leukemia or lymphoma from Europe and Australia—included 112 adults (and 166 children) with generally large tumor burdens, as assessed by WBC counts for leukemia and clinical stage and also by serum LDH levels for lymphoma.14 Reduction in PUA levels with rasburicase was statistically and clinically highly significant, resulting in PUA response rates of 100% in both adults and children. Only one adult patient, a 41-year-old man with AML and hyperuricemia at baseline, required hemodialysis because of acute renal failure, although PUA levels returned to normal after treatment with rasburicase. The patient subsequently died of respiratory failure attributable to disease ¬progression.14
The multicenter US registrational trial for the adult indication of rasburicase included 280 patients with hematologic malignancies who were at risk of TLS.55 Patients were randomly assigned to one of three treatments: rasburicase (0.2 mg/kg/d given as a 30-minute IV infusion) for 5 days, rasburicase for 3 days followed by oral allopurinol (300 mg/d) for 2 days, or allopurinol for 5 days. Rasburicase alone achieved significantly greater response rates than allopurinol monotherapy both in patients with a high risk of TLS and in those with baseline hyperuricemia (Table 2). In this clinical trial, control of PUA concentrations was achieved within 4 hours with rasburicase (compared with a median time interval of 27 hours with allopurinol) and was maintained during the 7-day monitoring period after initiation of antihyperuricemic therapy. Although reduction in the incidence of TLS was not an efficacy endpoint of the study, safety data analysis revealed that the occurrence of LTLS or CTLS was less common in the rasburicase than in the allopurinol treatment arm.55
In this large, randomized clinical study, rasburicase was safe and generally well tolerated during the 5-day treatment period. Patients who received rasburicase and those who received allopurinol had a similar incidence of adverse events.55,56 The only drug-related adverse events observed among the 184 patients who received rasburicase (alone or in combination with allopurinol) were potential hypersensitivity reactions in five patients, including irritation at the injection site (one patient); arthralgia, myalgia, and rash (one patient); peripheral edema (one patient); and grade 3 hypersensitivity (two patients). One patient with grade 3 hypersensitivity related to rasburicase discontinued study participation on day 1. No grade 4 hypersensitivity reaction, anaphylaxis, hemolytic reaction, or methemoglobinemia occurred with rasburicase.55
In summary, the results of this phase III randomized controlled study show that rasburicase may be superior to allopurinol in preventing hyperuricemia in adult leukemia/lymphoma patients at high risk of TLS and in achieving rapid and effective control of PUA levels in patients with hyperuricemia. The results support the recommendation within the current TLS management guidelines of using rasburicase for prophylaxis in high-risk patients and for treatment of hyperuricemia of malignancy, as well as in those patients with fully developed LTLS or CTLS (irrespective of PUA levels).17
Conclusion
In adult patients with cancer, the risk of serious clinical consequences of TLS often is increased by the presence of renal impairment, heart disease, baseline metabolic imbalances, and multiorgan derangements. Even in adult patients without serious comorbidities, an age-related decrease in physiologic resistance (reduced pathobiologic reserve), lifestyle factors (eg, adverse chronic exposure to tobacco, alcohol, or other toxins; sedentary existence; obesity; and vitamin and macro/micronutrient deficiencies), and dependence on multiple medications (polypharmacy and an associated increased risk for drug-drug interactions) should be considered for both TLS risk assessment and therapeutic choices in TLS prophylaxis and treatment. Although TLS is most common in patients with hematologic malignancies, serious cases of TLS have been reported in patients with a large variety of solid tumors, with sometimes fatal outcomes.
Assessment of TLS risk in patients with solid tumors, who generally tend to belong to a more aged population, is more difficult than in those with hematologic malignancies. Tumor burden and the type of chemotherapy are the most important risk factors for development of TLS in association with solid tumors; however, an unexpected occurrence of acute spontaneous TLS with hyperuricemia has been observed, even in the absence of these two factors.
Hyperuricemia is a key manifestation of TLS and is associated with risks of AKI, fluid overload, heart failure, and death. Prevention of hyperuricemia in high-risk patients and its rapid reversal in those with hyperuricemia at presentation or in those with fully developed TLS are of utmost importance in the successful management of TLS in adults with cancer. Results of the recent phase III study of rasburicase in adult patients with cancer demonstrated that rasburicase is superior to allopurinol in providing rapid and effective control of PUA levels.
Acknowledgments: Editorial assistance provided by Roland Tacke, PhD, and Candace Lundin, DVM, MS, was funded by sanofi-aventis US. The authors were fully responsible for all content and editorial decisions and did not receive financial support or compensation related to the development of this article.
References
- Cairo MS, Bishop M. Tumour lysis syndrome: new therapeutic strategies and classification. Br J Haematol 2004;127:3–11.
- Mughal TI, Ejaz AA, Foringer JR, Coiffier B. An integrated clinical approach for the identification, prevention, and treatment of tumor lysis syndrome. Cancer Treat Rev 2010;36:164–176.
- Hande KR, Garrow GC. Acute tumor lysis syndrome in patients with high-grade non-Hodgkin’s lymphoma. Am J Med 1993;94:133–139.
- Annemans L, Moeremans K, Lamotte M, et al. Incidence, medical resource utilisation and costs of hyperuricemia and tumour lysis syndrome in patients with acute leukaemia and non-Hodgkin’s lymphoma in four European countries. Leuk Lymphoma 2003;44:77–83.
- Coiffier B, Riouffol C. Management of tumor lysis syndrome in adults. Expert Rev Anticancer Ther 2007;7:233–239.
- Pumo V, Sciacca D, Malaquarnera M. Tumor lysis syndrome in elderly. Crit Rev ¬Oncol Hematol 2007;64:31–42.
- Kennedy LD, Ajiboye VO. Rasburicase for the prevention and treatment of hyperuricemia in tumor lysis syndrome. J Oncol Pharm Pract 2010;16:205–213.
- Allison SP, Lobo DN. Fluid and electrolytes in the elderly. Curr Opin Clin Nutr Metab Care 2004;7:27–33.
- Michelis MF. Hyperkalemia in the elderly. Am J Kidney Dis 1990;16:296–299.
- Williams ME. Endocrine crises: hyperkalemia. Crit Care Clin 1991;7:155–174.
- Weir MR. Non-diuretic-based antihypertensive therapy and potassium homeostasis in elderly patients. Coron Artery Dis 1997;8:499–504.
- Sica DA. Antihypertensive therapy and its effects on potassium homeostasis. J Clin Hypertens (Greenwich) 2006;8:67–73.
- Moe SM. Disorders involving calcium, phosphorus, and magnesium. Prim Care 2008;35:215–237.
- Bosly A, Sonet A, Pinkerton CR, et al. Rasburicase (recombinant urate oxidase) for the management of hyperuricemia in patients with cancer: report of an international compassionate use study. Cancer 2003;98:1048–1054.
- Jeha S, Kantarjian H, Irwin D, et al. Efficacy and safety of rasburicase, a recombinant urate oxidase (Elitek), in the management of malignancy-associated hyperuricemia in pediatric and adult patients: final results of a multicenter compassionate use trial. Leukemia 2005;19:34–38.
- Montesinos P, Lorenzo I, Martin G, et al. Tumor lysis syndrome in patients with acute myeloid leukemia: identification of risk factors and development of a predictive model. Haematologica 2008;93:67–74.
- Coiffier B, Altman A, Pui CH, Younes A, Cairo MS. Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review. J Clin Oncol 2008;26:2767–2778.
- Cairo MS, Coiffier B, Reiter A, Younes A; TLS Expert Panel. Recommendations for the evaluation of risk and prophylaxis of tumour lysis syndrome (TLS) in adults and children with malignant diseases: an expert TLS panel consensus. Br J Haematol 2010;149:578–586.
- Gemici C. Tumour lysis syndrome in solid tumours. Clin Oncol (R Coll Radiol) 2006;18:773–780.
- Pinder EM, Atwal GS, Ayantunde AA, et al. Tumour lysis syndrome occurring in a patient with metastatic gastrointestinal stromal tumour treated with Glivec (imatinib mesylate, Gleevec, STI571). Sarcoma 2007;2007:82012.
- Mott FE, Esana A, Chakmakjian C, Herrington JD. Tumor lysis syndrome in solid tumors. Support Cancer Ther 2005;2:188–191.
- Konuma T, Ooi J, Takahashi S, et al. Fatal acute tumor lysis syndrome following intrathecal chemotherapy for acute lymphoblastic leukemia with meningeal involvement. Intern Med 2008;47:1987–1988.
- Chang H, Shih LY. Imatinib-induced tumor lysis syndrome: report of a case and review of the literature. Chang Gung Med J 2008;31:510–514.
- Rüger W, Kruip MJ, Betjes MG. Reversible renal failure due to bilateral renal sarcoma in a patient with acute myeloid leukemia. Ren Fail 2009;31:606–609.
- Kosseifi SG, Vyas B, Vyas H, Krishnaswamy G, Smith S. Tumor lysis syndrome following chlorambucil therapy for chronic lymphocytic leukemia: case report and review of the literature. Tenn Med 2009;102:33–36.
- Calvo-Villas JM, Urcuyo BM, Umpierrez AM, Sicilia F. Acute tumor lysis syndrome during oral fludarabine treatment for chronic lymphocytic leukemia: role of treatment with rasburicase. Onkologie 2008;31:197–199.
- Al-Kali A, Farooq S, Tfayli A. Tumor lysis syndrome after starting treatment with Gleevec in a patient with chronic myelogenous leukemia. J Clin Pharm Ther 2009;34:607–610.
- Sonmez M, Ovali E, Omay SB. Tumor lysis syndrome during treatment with AMN107 (nilotinib) in a patient with chronic myelogenous leukemia accelerated phase. J Clin Pharm Ther 2008;33:91–92.
- Lin CJ, Chen HH, Hsieh RK, Chen YC, Wu CJ. Acute tumor lysis syndrome in a hemodialysis patient with diffuse large B cell lymphoma. Med Oncol 2009;26:93–95.
- Kasahara I, Nishio M, Yamamoto S, et al. Cord blood transplantation with a reduced-intensity conditioning regimen for patients with relapsed aggressive multiple myeloma after cytoreduction with bortezomib. Int J Hematol 2009;90:413–415.
- Chim CS. Rapid complete remission in multiple myeloma with bortezomib/thalidomide/dexamethasone combination therapy following development of tumor lysis syndrome. Cancer Chemother Pharmacol 2008;62:181–182.
- Bouaziz JD, Cordel N, Hickman G, Fieschi C, Ortonne N, Bagot M. Cutaneous tumor lysis syndrome in a patient with HTLV-1 adult T-cell lymphoma/leukemia. Blood 2009;114:4320–4321.
- Fritsch-Stork RD, Leguit RJ, Derksen RH. Rapidly fatal HTLV-1-associated T-cell leukemia/lymphoma in a patient with SLE. Nat Rev Rheumatol 2009;5:283–287.
- Hentrich M, Schiel X, Scheidt B, Reitmeier M, Hoffmann U, Lutz L. Fatal tumor lysis syndrome after irinotecan/5-FU/folinic acid/bevacizumab-containing therapy in a patient heavily pretreated for metastatic colon cancer. Acta Oncol 2008;47:155–156.
- Feres GA, Salluh JI, Ferreira CG, Soares M. Severe acute tumor lysis syndrome in patients with germ-cell tumors. Indian J Urol 2008;24:555–557.
- Hsieh PM, Hung KC, Chen YS. Tumor lysis syndrome after transarterial chemoembolization of hepatocellular carcinoma: case reports and literature review. World J Gastroenterol 2009;15:4726–4728.
- Huang WS, Yang CH. Sorafenib induced tumor lysis syndrome in an advanced hepatocellular carcinoma patient. World J Gastroenterol 2009;15:4464–4466.
- Noh GY, Choe DH, Kim CH, Lee JC. Fatal tumor lysis syndrome during radiotherapy for non-small-cell lung cancer. J Clin Oncol 2008;26:6005–6006.
- Froilán Torres C, Castro Carbajo P, Pajares Villarroya R, et al. Acute spontaneous tumor lysis syndrome in a patient with Crohn’s disease taking immunosuppressants. Rev Esp Enferm Dig 2009;101:288–294.
- Michels J, Lassau N, Gross-Goupil M, Massard C, Mejean A, Escudier B. Sunitinib inducing tumor lysis syndrome in a patient treated for renal carcinoma. Invest New Drugs 2010;28:690–693.
- Shenoy C. Acute spontaneous tumor lysis syndrome in a patient with squamous cell carcinoma of the lung. QJM 2009;102:71–73.
- Nakamura Y, Nakamura Y, Hori E, et al. Tumor lysis syndrome after transcatheter arterial infusion of cisplatin and embolization therapy for liver metastases of melanoma. Int J Dermatol 2009;48:763–767.
- Yamazaki H, Hanada M, Horiki M, et al. Acute tumor lysis syndrome caused by palliative radiotherapy in patients with diffuse large B-cell lymphoma. Radiat Med 2004;22:52–55.
- Cheson BD. Etiology and management of tumor lysis syndrome in patients with chronic lymphocytic leukemia. Clin Adv Hematol Oncol 2009;7:263–271.
- Lin TS. Novel agents in chronic lymphocytic leukemia: efficacy and tolerability of new therapies. Clin Lymphoma Myeloma 2008;8(suppl 4):S137–S143.
- Rituxan [package insert]. South San Francisco, CA: Genentech, Inc.; 2011.
- Treanda [package insert]. Frazer, PA: Cephalon, Inc.; 2010.
- Lin TS, Ruppert AS, Johnson AJ, et al. Phase II study of flavopiridol in relapsed chronic lymphocytic leukemia demonstrating high response rates in genetically high-risk disease. J Clin Oncol 2009;27:6012–6018.
- Byrd JC, Lin TS, Dalton JT, et al. Flavopiridol administered using a pharmacologically derived schedule is associated with marked clinical efficacy in refractory, genetically high-risk chronic lymphocytic leukemia. Blood 2007;109:399–404.
- Moutouh-de Parseval LA, Weiss L, DeLap RJ, Knight RD, Zeldis JB. Tumor lysis syndrome/tumor flare reaction in lenalidomide-treated chronic lymphocytic leukemia. J Clin Oncol 2007;25:5047.
- Sezer O, Vesole DH, Singhal S, et al. Bortezomib-induced tumor lysis syndrome in multiple myeloma. Clin Lymphoma Myeloma 2006;7:233–235.
- Huston A, Brown J, Roodman GD. Tumor lysis syndrome following thalidomide and dexamethasone therapy for newly diagnosed multiple myeloma. Exp Hematol 2006;34:1616.
- Berenson JR, Yang HH, Vescio RA, et al. Safety and efficacy of bortezomib and melphalan combination in patients with relapsed or refractory multiple myeloma: updated results of a phase 1/2 study after longer follow-up. Ann Hematol 2008;87:623–631.
- Fuente N, Mañe JM, Barcelo R, Muñoz A, Perez-Hoyos T, Lopez-Vivanco G. Tumor lysis syndrome in a multiple myeloma patient treated with thalidomide. Ann Oncol 2004;15:537.
- Cortes J, Moore JO, Maziarz RT, et al. Control of plasma uric acid in adults at risk for tumor lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone: results of a multicenter phase III study. J Clin Oncol 2010;28:4207–4213.
- Elitek [package insert]. Bridgewater, NJ: sanofi-aventis U.S., LLC; 2009.
ABOUT THE AUTHORS
Affiliations: Dr. Seiter is Professor of Medicine and Director of the Leukemia Service at New York Medical College, Valhalla, NY. Dr. Sarlis is currently Vice President and Head of the Medical Affairs Department at Incyte Corporation, Wilmington, DE. Dr. Kim is Associate Professor and Chief, Section of Head and Neck Medical Oncology, Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, at The University of Texas MD Anderson Cancer Center, Houston, TX.
Conflicts of interest: Dr. Sarlis was an employee of sanofi-aventis U.S. at the time this article was written and holds stock options and stock in this company. Drs. Seiter and Kim have no pertinent conflicts of interest to disclose.
Karen P. Seiter, MD,1 Nicholas J. Sarlis, MD, PhD, FACP,2* and Edward S. Kim, MD3
- New York Medical College, Valhalla, NY;
- Medical Affairs–Oncology, sanofi-aventis U.S., Bridgewater, NJ; and
- The University of Texas MD Anderson Cancer Center, Houston, TX
Government audits: ‘big brother’ is monitoring you
What does an oncologist need to know about OIGs, RACs, and ZPICs? Plenty. They are some of the auditors who can make life difficult if a practice is not in compliance with Medicare and Medicaid billing and utilization procedures, Roberta L. Buell said at this journal’s annual Community Oncology Conference in Las Vegas.
The government is financing health¬care reform in part through audits to recover stolen and misused money, she explained. Thus far, it has recovered impressive sums, suggesting that the auditors are here to stay no matter how reform plays out. Anyone who runs a practice could be audited, cautioned Ms. Buell, a principal and coding and reimbursement specialist at onPoint Oncology in Sausalito, California. She discussed some of the key types of auditors and their roles.
Acronyms with a mission
Office of Inspector General auditors (OIGs) are responsible for ensuring appropriate use of Medicare funds. Their activities range from conducting basic audits to arranging arrests if a practice is deemed to have done something criminal.
OIGs will be scrutinizing a host of issues in 2011 and 2012, according to Ms. Buell. One is “place-of-service” errors, whereby a claim indicates a patient was treated in an office, where reimbursement is higher, but treatment actually took place in a hospital or ambulatory surgical center. This can be tricky for oncologists who sell their practices to a hospital but maintain a separate office. When physicians see patients in the office, “they’re…billing Part B. But if they walk across the hall to the infusion center and they see [patients] over there,…their place of service will be the hospital,” she said.
The OIGs will also be taking a close look at evaluation and management (E/M) codes to assess whether the codes accurately reflect the type, setting, and complexity of services provided as well as patient status—new or established. Also within the E/M category, auditors will be on the look out for identical (boilerplate) documentation across patients, sometimes a result of use of electronic medical records.
Error-prone providers, such as those with high numbers of duplicate claims, will catch the auditors’ attention as well. To avoid this, Ms. Buell recommended following up with a phone call when a claim goes unpaid, rather than resubmitting the claim. Medicare incentive payments for use of electronic medical records will also feature in the auditors’ work plan. “People are going to be randomly audited to make sure that they had 90 days of consecutive meaningful use—and it has to be for all patients, not just for Medicare patients,” Ms. Buell said.
Compliance with the Health Insurance Portability and Accountability Act will also be assessed. Some large fines have already been levied because of data-security breaches, so practices need to be especially vigilant about not leaving records around or losing laptops.
The OIGs will also scrutinize a variety of issues related to the Part D prescription drug program, such as inclusion of Part A and B claims with Part D claims. Ms. Buell cited billing practices for erythropoiesis-stimulating agents and pegfilgrastim (Neulasta) as an example: a practice might bill through Part D for Neulasta because it is “under water” on it, but “that is going to come back to haunt [you], because that’s not supposed to happen.”
A related aspect of Part D billing is the duplication of drug claims for hospice patients. “If somebody is in the hospice and is getting a drug through the hospice, but comes to your practice and gets it again, that’s going to be looked at,” she explained.
Zone Program Integrity Contractors (ZPICs) “are the [auditors] you really have to worry about. These are the folks who are looking for fraud.” Their data-analysis program is intended to identify provider billing practices and services that pose the greatest financial risk to the Medicare program, for example, high-volume and high-cost services that are being widely overused. But they also investigate credible whistle-blower (qui tam) complaints.
ZPICs can send a practice two types of letters. One is a request for records within 30 days, which usually means the practice was flagged by the screening program and the auditors want to take a closer look at the documentation. In this case, an attorney is not necessary. “But you have to have people look very hard at those records…[so] that everything that is done is justified in the record,” she said. The other type of letter usually arrives by fax and states that auditors will be on site within a few days. “That means that they have a complaint, either by a patient or more likely by a disgruntled physician or employee,” Ms. Buell noted. Here, the stakes are criminal penalties, and there should be an attorney on site during the visit.
Recovery Audit Contractors (RACs) “are bounty hunters,” she said. They are focused on recovering misspent Medicare funds and are paid a contingency fee of 9%–12% of the amount recovered. However, they are not active in practices because they have enjoyed such success in hospitals. At present, they do automated screening of practices’ Part B payments. “If you fall out of the screen, they ask for the money back and that’s the end,” Ms. Buell said. She cautioned, however, that RACs might start doing complex reviews later this year. And if they do, drugs are likely to be an early focus “because there is so much money involved and they have seen that [in the hospitals].”
Medicaid Integrity Contractors (MICs) review Medicaid claims for inappropriate payments and fraud, using a data-driven approach to identify aberrant billing practices. Unlike RACs, who receive a contingency fee, MICs are paid for their services and receive a quality-related bonus. Their look-back period for medical records varies by state. At present, they target mainly hospitals and, to a lesser extent, skilled nursing facilities, nursing homes, and hospices. Physicians are low on the list of priorities, “but…if you have a heavy Medicaid load, you do have to worry about [it],” Ms. Buell said.
Medicare Administrative Contractors (MACs) process claims for both Part A and Part B services and therefore can review discrepancies between the two sets of claims, revise payments, and increase denials. Some ongoing MAC activities include audits regarding usage of the 99204 and 99205 procedure codes for new patients.
Minimizing the risks
Given this environment, what can oncology practices do to minimize their risk? “Have a compliance [program] and make it a priority,” Ms. Buell recommended, adding that it is also a requirement of healthcare reform. Be sure to fix anything the program identifies, because auditors will take a much stiffer stance if they discover issues that were known but not resolved.
Practices can refer to a set of components that the OIG has set forth as the foundation of an effective compliance and ethics program (www.oig.hhs.gov/authorities/docs/physician.pdf). For example, there should be standard policies and procedures in the practice for things such as documentation, education and training, and updates on Medicare regulations. The enforcement of these policies is also a key element of an effective compliance program, as is a prevention component that anticipates mistakes and prevents them from occurring whenever possible.
The healthcare reform legislation requires that practices report and return Medicare and Medicaid overpayments. “If you find that you have been overpaid, for any reason, even if it’s their mistake, give the money back,” Ms. Buell said, because there is a good chance that auditors will eventually discover it.
Any practice that is concerned about being at risk for whistle-blower incidents should obtain legal help. “If you think there is…someone out there who can hurt you, you need to have your compliance program administered by an attorney.”
Finally, “educate, educate, educate,” she advised, to keep physicians and other practice members current on compliance issues.
What does an oncologist need to know about OIGs, RACs, and ZPICs? Plenty. They are some of the auditors who can make life difficult if a practice is not in compliance with Medicare and Medicaid billing and utilization procedures, Roberta L. Buell said at this journal’s annual Community Oncology Conference in Las Vegas.
The government is financing health¬care reform in part through audits to recover stolen and misused money, she explained. Thus far, it has recovered impressive sums, suggesting that the auditors are here to stay no matter how reform plays out. Anyone who runs a practice could be audited, cautioned Ms. Buell, a principal and coding and reimbursement specialist at onPoint Oncology in Sausalito, California. She discussed some of the key types of auditors and their roles.
Acronyms with a mission
Office of Inspector General auditors (OIGs) are responsible for ensuring appropriate use of Medicare funds. Their activities range from conducting basic audits to arranging arrests if a practice is deemed to have done something criminal.
OIGs will be scrutinizing a host of issues in 2011 and 2012, according to Ms. Buell. One is “place-of-service” errors, whereby a claim indicates a patient was treated in an office, where reimbursement is higher, but treatment actually took place in a hospital or ambulatory surgical center. This can be tricky for oncologists who sell their practices to a hospital but maintain a separate office. When physicians see patients in the office, “they’re…billing Part B. But if they walk across the hall to the infusion center and they see [patients] over there,…their place of service will be the hospital,” she said.
The OIGs will also be taking a close look at evaluation and management (E/M) codes to assess whether the codes accurately reflect the type, setting, and complexity of services provided as well as patient status—new or established. Also within the E/M category, auditors will be on the look out for identical (boilerplate) documentation across patients, sometimes a result of use of electronic medical records.
Error-prone providers, such as those with high numbers of duplicate claims, will catch the auditors’ attention as well. To avoid this, Ms. Buell recommended following up with a phone call when a claim goes unpaid, rather than resubmitting the claim. Medicare incentive payments for use of electronic medical records will also feature in the auditors’ work plan. “People are going to be randomly audited to make sure that they had 90 days of consecutive meaningful use—and it has to be for all patients, not just for Medicare patients,” Ms. Buell said.
Compliance with the Health Insurance Portability and Accountability Act will also be assessed. Some large fines have already been levied because of data-security breaches, so practices need to be especially vigilant about not leaving records around or losing laptops.
The OIGs will also scrutinize a variety of issues related to the Part D prescription drug program, such as inclusion of Part A and B claims with Part D claims. Ms. Buell cited billing practices for erythropoiesis-stimulating agents and pegfilgrastim (Neulasta) as an example: a practice might bill through Part D for Neulasta because it is “under water” on it, but “that is going to come back to haunt [you], because that’s not supposed to happen.”
A related aspect of Part D billing is the duplication of drug claims for hospice patients. “If somebody is in the hospice and is getting a drug through the hospice, but comes to your practice and gets it again, that’s going to be looked at,” she explained.
Zone Program Integrity Contractors (ZPICs) “are the [auditors] you really have to worry about. These are the folks who are looking for fraud.” Their data-analysis program is intended to identify provider billing practices and services that pose the greatest financial risk to the Medicare program, for example, high-volume and high-cost services that are being widely overused. But they also investigate credible whistle-blower (qui tam) complaints.
ZPICs can send a practice two types of letters. One is a request for records within 30 days, which usually means the practice was flagged by the screening program and the auditors want to take a closer look at the documentation. In this case, an attorney is not necessary. “But you have to have people look very hard at those records…[so] that everything that is done is justified in the record,” she said. The other type of letter usually arrives by fax and states that auditors will be on site within a few days. “That means that they have a complaint, either by a patient or more likely by a disgruntled physician or employee,” Ms. Buell noted. Here, the stakes are criminal penalties, and there should be an attorney on site during the visit.
Recovery Audit Contractors (RACs) “are bounty hunters,” she said. They are focused on recovering misspent Medicare funds and are paid a contingency fee of 9%–12% of the amount recovered. However, they are not active in practices because they have enjoyed such success in hospitals. At present, they do automated screening of practices’ Part B payments. “If you fall out of the screen, they ask for the money back and that’s the end,” Ms. Buell said. She cautioned, however, that RACs might start doing complex reviews later this year. And if they do, drugs are likely to be an early focus “because there is so much money involved and they have seen that [in the hospitals].”
Medicaid Integrity Contractors (MICs) review Medicaid claims for inappropriate payments and fraud, using a data-driven approach to identify aberrant billing practices. Unlike RACs, who receive a contingency fee, MICs are paid for their services and receive a quality-related bonus. Their look-back period for medical records varies by state. At present, they target mainly hospitals and, to a lesser extent, skilled nursing facilities, nursing homes, and hospices. Physicians are low on the list of priorities, “but…if you have a heavy Medicaid load, you do have to worry about [it],” Ms. Buell said.
Medicare Administrative Contractors (MACs) process claims for both Part A and Part B services and therefore can review discrepancies between the two sets of claims, revise payments, and increase denials. Some ongoing MAC activities include audits regarding usage of the 99204 and 99205 procedure codes for new patients.
Minimizing the risks
Given this environment, what can oncology practices do to minimize their risk? “Have a compliance [program] and make it a priority,” Ms. Buell recommended, adding that it is also a requirement of healthcare reform. Be sure to fix anything the program identifies, because auditors will take a much stiffer stance if they discover issues that were known but not resolved.
Practices can refer to a set of components that the OIG has set forth as the foundation of an effective compliance and ethics program (www.oig.hhs.gov/authorities/docs/physician.pdf). For example, there should be standard policies and procedures in the practice for things such as documentation, education and training, and updates on Medicare regulations. The enforcement of these policies is also a key element of an effective compliance program, as is a prevention component that anticipates mistakes and prevents them from occurring whenever possible.
The healthcare reform legislation requires that practices report and return Medicare and Medicaid overpayments. “If you find that you have been overpaid, for any reason, even if it’s their mistake, give the money back,” Ms. Buell said, because there is a good chance that auditors will eventually discover it.
Any practice that is concerned about being at risk for whistle-blower incidents should obtain legal help. “If you think there is…someone out there who can hurt you, you need to have your compliance program administered by an attorney.”
Finally, “educate, educate, educate,” she advised, to keep physicians and other practice members current on compliance issues.
What does an oncologist need to know about OIGs, RACs, and ZPICs? Plenty. They are some of the auditors who can make life difficult if a practice is not in compliance with Medicare and Medicaid billing and utilization procedures, Roberta L. Buell said at this journal’s annual Community Oncology Conference in Las Vegas.
The government is financing health¬care reform in part through audits to recover stolen and misused money, she explained. Thus far, it has recovered impressive sums, suggesting that the auditors are here to stay no matter how reform plays out. Anyone who runs a practice could be audited, cautioned Ms. Buell, a principal and coding and reimbursement specialist at onPoint Oncology in Sausalito, California. She discussed some of the key types of auditors and their roles.
Acronyms with a mission
Office of Inspector General auditors (OIGs) are responsible for ensuring appropriate use of Medicare funds. Their activities range from conducting basic audits to arranging arrests if a practice is deemed to have done something criminal.
OIGs will be scrutinizing a host of issues in 2011 and 2012, according to Ms. Buell. One is “place-of-service” errors, whereby a claim indicates a patient was treated in an office, where reimbursement is higher, but treatment actually took place in a hospital or ambulatory surgical center. This can be tricky for oncologists who sell their practices to a hospital but maintain a separate office. When physicians see patients in the office, “they’re…billing Part B. But if they walk across the hall to the infusion center and they see [patients] over there,…their place of service will be the hospital,” she said.
The OIGs will also be taking a close look at evaluation and management (E/M) codes to assess whether the codes accurately reflect the type, setting, and complexity of services provided as well as patient status—new or established. Also within the E/M category, auditors will be on the look out for identical (boilerplate) documentation across patients, sometimes a result of use of electronic medical records.
Error-prone providers, such as those with high numbers of duplicate claims, will catch the auditors’ attention as well. To avoid this, Ms. Buell recommended following up with a phone call when a claim goes unpaid, rather than resubmitting the claim. Medicare incentive payments for use of electronic medical records will also feature in the auditors’ work plan. “People are going to be randomly audited to make sure that they had 90 days of consecutive meaningful use—and it has to be for all patients, not just for Medicare patients,” Ms. Buell said.
Compliance with the Health Insurance Portability and Accountability Act will also be assessed. Some large fines have already been levied because of data-security breaches, so practices need to be especially vigilant about not leaving records around or losing laptops.
The OIGs will also scrutinize a variety of issues related to the Part D prescription drug program, such as inclusion of Part A and B claims with Part D claims. Ms. Buell cited billing practices for erythropoiesis-stimulating agents and pegfilgrastim (Neulasta) as an example: a practice might bill through Part D for Neulasta because it is “under water” on it, but “that is going to come back to haunt [you], because that’s not supposed to happen.”
A related aspect of Part D billing is the duplication of drug claims for hospice patients. “If somebody is in the hospice and is getting a drug through the hospice, but comes to your practice and gets it again, that’s going to be looked at,” she explained.
Zone Program Integrity Contractors (ZPICs) “are the [auditors] you really have to worry about. These are the folks who are looking for fraud.” Their data-analysis program is intended to identify provider billing practices and services that pose the greatest financial risk to the Medicare program, for example, high-volume and high-cost services that are being widely overused. But they also investigate credible whistle-blower (qui tam) complaints.
ZPICs can send a practice two types of letters. One is a request for records within 30 days, which usually means the practice was flagged by the screening program and the auditors want to take a closer look at the documentation. In this case, an attorney is not necessary. “But you have to have people look very hard at those records…[so] that everything that is done is justified in the record,” she said. The other type of letter usually arrives by fax and states that auditors will be on site within a few days. “That means that they have a complaint, either by a patient or more likely by a disgruntled physician or employee,” Ms. Buell noted. Here, the stakes are criminal penalties, and there should be an attorney on site during the visit.
Recovery Audit Contractors (RACs) “are bounty hunters,” she said. They are focused on recovering misspent Medicare funds and are paid a contingency fee of 9%–12% of the amount recovered. However, they are not active in practices because they have enjoyed such success in hospitals. At present, they do automated screening of practices’ Part B payments. “If you fall out of the screen, they ask for the money back and that’s the end,” Ms. Buell said. She cautioned, however, that RACs might start doing complex reviews later this year. And if they do, drugs are likely to be an early focus “because there is so much money involved and they have seen that [in the hospitals].”
Medicaid Integrity Contractors (MICs) review Medicaid claims for inappropriate payments and fraud, using a data-driven approach to identify aberrant billing practices. Unlike RACs, who receive a contingency fee, MICs are paid for their services and receive a quality-related bonus. Their look-back period for medical records varies by state. At present, they target mainly hospitals and, to a lesser extent, skilled nursing facilities, nursing homes, and hospices. Physicians are low on the list of priorities, “but…if you have a heavy Medicaid load, you do have to worry about [it],” Ms. Buell said.
Medicare Administrative Contractors (MACs) process claims for both Part A and Part B services and therefore can review discrepancies between the two sets of claims, revise payments, and increase denials. Some ongoing MAC activities include audits regarding usage of the 99204 and 99205 procedure codes for new patients.
Minimizing the risks
Given this environment, what can oncology practices do to minimize their risk? “Have a compliance [program] and make it a priority,” Ms. Buell recommended, adding that it is also a requirement of healthcare reform. Be sure to fix anything the program identifies, because auditors will take a much stiffer stance if they discover issues that were known but not resolved.
Practices can refer to a set of components that the OIG has set forth as the foundation of an effective compliance and ethics program (www.oig.hhs.gov/authorities/docs/physician.pdf). For example, there should be standard policies and procedures in the practice for things such as documentation, education and training, and updates on Medicare regulations. The enforcement of these policies is also a key element of an effective compliance program, as is a prevention component that anticipates mistakes and prevents them from occurring whenever possible.
The healthcare reform legislation requires that practices report and return Medicare and Medicaid overpayments. “If you find that you have been overpaid, for any reason, even if it’s their mistake, give the money back,” Ms. Buell said, because there is a good chance that auditors will eventually discover it.
Any practice that is concerned about being at risk for whistle-blower incidents should obtain legal help. “If you think there is…someone out there who can hurt you, you need to have your compliance program administered by an attorney.”
Finally, “educate, educate, educate,” she advised, to keep physicians and other practice members current on compliance issues.
Feasibility and acceptance of a telehealth intervention to promote symptom management during treatment for head and neck cancer
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article |
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article |
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article |
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article |
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article |
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article |
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract |
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article |
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article |
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article |
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article |
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article |
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article |
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article |
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract |
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article |
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article |
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article |
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article |
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article |
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article |
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article |
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract |
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article |
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
Results of a Multicenter Open-Label Randomized Trial Evaluating Infusion Duration of Zoledronic Acid in Multiple Myeloma Patients (the ZMAX Trial)
Original research
James R. Berenson MD
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article |
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.
Original research
James R. Berenson MD
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article |
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.
Original research
James R. Berenson MD
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article |
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.