Multiple myeloma (MM) typically afflicts elderly patients with a median age of 65 years. However, while recently shown to provide superior outcome to standard treatment, high-dose therapy (HDT) has usually been limited to patients up to 65 years. Among 550 patients with MM and a minimum follow-up of 18 months, 49 aged ≥65 years were identified (median age, 67; range, 65 to 76 years). Their outcome was compared with 49 younger pair mates (median, 52; range, 37 to 64 years) selected among the remaining 501 younger patients (<65 years) matched for five previously recognized critical prognostic factors (cytogenetics, β2-microglobulin, C-reactive protein, albumin, creatinine). Nearly one half had been treated for more than 1 year with standard therapy and about one third had refractory MM. All patients received high-dose melphalan-based therapy; 76% of the younger and 65% of the older group completed a second transplant (P = .3). Sufficient peripheral blood stem cells to support two HDT cycles (CD34 > 5 × 106/kg) were available in 83% of younger and 73% of older patients (P = .2). After HDT, hematopoietic recovery to critical levels of granulocytes (>500/μL) and of platelets (>50,000/μL) proceeded at comparable rates among younger and older subjects with both first and second HDT. The frequency of extramedullary toxicities was comparable. Treatment-related mortality with the first HDT cycle was 2% in younger and 8% among older subjects, whereas no mortality was encountered with the second transplant procedure. Comparing younger/older subjects, median durations of event-free and overall survival were 2.8/1.5 years (P = .2) and 4.8/3.3 years (P = .4). Multivariate analysis showed pretransplant cytogenetics and β2-microglobulin levels as critical prognostic features for both event-free and overall survival, whereas age was insignificant for both endpoints (P = .2/.8). Thus, age is not a biologically adverse parameter for patients with MM receiving high-dose melphalan-based therapy with peripheral blood stem cell support and, hence, should not constitute an exclusion criterion for participation in what appears to be superior therapy for symptomatic MM.

MULTIPLE MYELOMA (MM) afflicts approximately 13,000 Americans annually. Symptom manifestations relate to anemia, osteopenia and lytic bone disease, hypercalcemia, renal failure, and immunodifficiency.1,2 Over one half of the patients are older than 65 years at diagnosis. After lack of progress with standard-dose therapy over three decades,2hematopoietic stem cell supported high-dose therapy (HDT) has become the treatment of choice for symptomatic patients, especially younger individuals who are thought to be able to withstand the potentially greater toxicity incurred with myeloablative regimens.3-5Fortunately, treatment-related mortality has decreased to 5% or less, mainly as a result of rapid hematopoietic engraftment with the use of peripheral blood stem cells (PBSC) collected early in the disease course and mobilized with hematopoeitic growth factors alone or in conjunction with chemotherapy.6 

Since the introduction of autotransplants for MM,7,8 our patients were eligible for HDT up to age 70. In recent years, we essentially discontinued an upper age limit when disease severity was judged to outweigh the anticipated toxicities from HDT. In extensive analyses of both newly diagnosed and previously treated patients, age did not factor in as an adverse feature for event-free survival (EFS) and overall survival (OS).9 Instead, prognosis was dominantly impacted by cytogenetics, β2-microglobulin (B2M), and C-reactive protein (CRP) as well as duration of prior therapy.10,11 

Prompted by health insurance considerations for patients 65 years and older, we examined patients’ outcome after autotransplant-supported HDT in relationship to age. This report compares our experience with HDT in 49 patients ≥65 years and 49 younger patients, after matching for prognostically relevant pretreatment features.

Between December 1989 and August 1997, 900 patients were enrolled in tandem autologous transplant trials using melphalan 200 mg/m2 (MEL200) for the first cycle.9 A second HDT cycle was usually administered within 3 to 6 months consisting of MEL200 in case partial response (PR) was sustained. Patients with less than PR status were offered combination therapy (MEL140 + total body irradiation [TBI] 850 to 1125 cGy) or MEL200 and added high-dose cyclophosphamide (HDCTX; usually 6 g/m2); the latter regimen was used when patients were deemed not to be candidates for a TBI-containing regimen because of prior local radiation. All PBSC were collected before both autotransplants, either with HDCTX plus granulocyte colony-stimulating factor (G-CSF) or granulocyte macrophage CSF (GM-CSF) or with G-CSF alone as reported previously.12,13 

Forty-nine patients aged ≥65 years were identified who had a minimum posttransplant follow-up of 18 months. During this time interval, 501 younger patients (<65 years) were also treated. Using a standardized Euclidian distance measure,14 49 pair mates were identified whose prognostically relevant disease, host, and treatment features were comparable (Table 1). Pair mates were established using the following pretransplant prognostic factors: B2M, albumin, creatinine, CRP, and the presence or absence of unfavorable chromosomal abnormalities (11q breakpoints, monosomy 13 or deletion 13q, and any translocation).10,11 

Table 1.

Comparability of Prognostic Features Among Young and Old Patients

Favorable Parameter
% Patients P
<65 yr (N = 49) 65-69 yr (N = 39) ≥70 yr (N = 10)
Cytogenetics  74  64 90  .2  
B2M ≤2.5 mg/L  51  33  40  .2  
CRP ≤4.0 mg/L  53  38  60  .3  
≤12 mo of prior therapy 55  54  70  .6  
Non-IgA isotype  82  85  70 .6  
Sensitive  59  64  70  .8  
LDH ≤190 U/L 88  92  80  .5  
Stage <III  49  58  60 .6  
Albumin >3.5 g/dL  51  46  40  .8 
Creatinine ≤2 mg/dL  96  95  100  .8  
Second transplant  76  69  50  .3  
CD34 >5 × 106/kg  83  72  80  .4 
Favorable Parameter
% Patients P
<65 yr (N = 49) 65-69 yr (N = 39) ≥70 yr (N = 10)
Cytogenetics  74  64 90  .2  
B2M ≤2.5 mg/L  51  33  40  .2  
CRP ≤4.0 mg/L  53  38  60  .3  
≤12 mo of prior therapy 55  54  70  .6  
Non-IgA isotype  82  85  70 .6  
Sensitive  59  64  70  .8  
LDH ≤190 U/L 88  92  80  .5  
Stage <III  49  58  60 .6  
Albumin >3.5 g/dL  51  46  40  .8 
Creatinine ≤2 mg/dL  96  95  100  .8  
Second transplant  76  69  50  .3  
CD34 >5 × 106/kg  83  72  80  .4 

All patients had to sign an informed consent indicating the potential benefit and toxicities associated with the tandem autotransplant programs. Protocols and consent forms had been reviewed and approved by the institutional review board and, where appropriate, by the National Cancer Institute and the Food and Drug Administration. Clinical endpoints considered treatment-related mortality (TRM) within 60 days of HDT, separately for first and second autotransplant. Additionally, TRM was computed 6 and 12 months after the first HDT cycle. Complete response (CR) was defined as the absence, on immunofixation analysis of serum and urine, of monoclonal protein in the presence of normal morphologic examination of bone marrow aspirate and biopsy with less than 1% of tumor cells identified on the basis of DNA-cytoplasmic immunoglobulin flow cytometric analysis.15 CR had to be documented on at least two occasions at a minimum time interval of 3 months. EFS and OS were computed from the first HDT cycle. CR duration was measured from the first onset of CR to disease relapse or death. In addition, hematopoietic recovery was determined using threshold levels for granulocytes of at least 500/μL and of platelets of at least 50,000/μL, both with first and second transplant. Grade III and IV extramedullary toxicities were also determined.

Statistical methods used chi-squared tests for comparison of patient characteristics, the Kaplan-Meier product limit method for estimation of survival,16 and the log-rank test for comparison for CR duration, EFS, and OS.17 Multivariate analysis was used to determine the prognostically independent variables associated with various clinical endpoints.18 

The median age in the younger group ranged from 37 to 64 years (median, 52 years) and from 65 to 76 years (median, 67 years; 70 years, 7 patients; 71 years, 1; 72 years, 1; 76 years, 1 patient) among the older patients (P = .0001). Patient characteristics were well matched for the main risk factors previously identified to affect EFS and OS after autotransplants10,11 (Table 1). Specifically, the incidence of favorable cytogenetics (absence of aberrations of chromosomes 11 and 13 as well as any translocation) was similar in young patients and in the two older cohorts. Fifty-five percent to 70% had ≥12 months of prior therapy. Fewer older patients presented with low B2M ≤2.5 mg/L (P = .2). The proportion of patients with high CD34 counts (>5 × 106/kg) was similar (P = .4). A second transplant was applied in 76% of patients <65 years, 69% in the 65 to 69 years group, and in 50% of those ≥70 years. As far as regimens were concerned, all patients received MEL200 with the first autograft. The second transplant consisted of MEL200 in 18 of 37 (49%) among those <65 years, 11 of 27 (41%) in the middle age group, and in 4 of 5 (80%) in the old age group. MEL140 + TBI/MEL-HDCTX/other regimens were applied in 8/5/6 patients <65 years; and 12/2/2 of those aged 65 to 69 years and 1/0/0 in the ≥70-year group.

Granulocyte and platelet recoveries were superimposable in both age groups, after first and second HDT cycle, indicating excellent PBSC quantities and function (Fig 1). The frequencies of extramedullary toxicities were similar in younger and older patients during both transplant procedures (Table2). TRM within 60 days of the first transplant was 2% in younger (intracranial hemorrhage, 1 patient) and 8% in older patients (death of unknown cause after complete hematopoietic recovery, 66 years; pneumonia, 69 years; intracranial hemorrhage subsequent to trauma, 70 years; sepsis caused by delayed engraftment after CD34-selected autograft, 70 years) (P = .2). None of the second transplant recipients in either age group experienced TRM.

Fig. 1.

Similar recovery kinetics of granulocytes (>500/μL) and of platelets (>50,000/μL) in young and old patients, with both first and second transplant. The fraction of patients with CD34 >5 × 106/kg was comparable between age groups.

Fig. 1.

Similar recovery kinetics of granulocytes (>500/μL) and of platelets (>50,000/μL) in young and old patients, with both first and second transplant. The fraction of patients with CD34 >5 × 106/kg was comparable between age groups.

Close modal
Table 2.

Grade >III Nonhematologic Toxicities

Toxicity 1st TransplantP2nd Transplant P
<65 yr (N = 49) ≥65 yr (N = 49) <65 yr (N = 37) ≥65 yr (N = 32)
Mucositis/diarrhea 20* 31  .2  19  34  .1  
Pneumonia/sepsis 24  12  .2  19  9  .3  
Early deaths  2  .2  0  0  — 
Toxicity 1st TransplantP2nd Transplant P
<65 yr (N = 49) ≥65 yr (N = 49) <65 yr (N = 37) ≥65 yr (N = 32)
Mucositis/diarrhea 20* 31  .2  19  34  .1  
Pneumonia/sepsis 24  12  .2  19  9  .3  
Early deaths  2  .2  0  0  — 
*

Percent of patients.

The incidence of CR was lower in older than in younger subjects (20v 43%, P = .02). EFS and OS durations were comparable (Fig 2) as were CR durations (Fig 3). Multivariate analysis of pretransplant prognostic variables among all 98 patients identified favorable cytogenetics and low B2M as good risk features for both EFS and OS; in addition, months of prior therapy was also important for EFS. However, age was not a significant risk factor for either EFS or OS once these variables were accounted for (Table3).

Fig. 2.

Similar durations of EFS and OS after autotransplant-supported HDT in patients <65 and ≥65 years.

Fig. 2.

Similar durations of EFS and OS after autotransplant-supported HDT in patients <65 and ≥65 years.

Close modal
Fig. 3.

Comparable durations of complete remission regardless of age.

Fig. 3.

Comparable durations of complete remission regardless of age.

Close modal
Table 3.

Age Not Important for Clinical Outcome on Multivariate Analysis

EFS POS P
Favorable cytogenetics .004  Favorable cytogenetics  .009  
≤12 mo prior therapy  .01  B2M ≤2.5 mg/L  .03  
B2M ≤2.5 mg/L  .01  ≤12 mo prior therapy  .4  
Age <65 yr  .2 Age <65 yr  .8 
EFS POS P
Favorable cytogenetics .004  Favorable cytogenetics  .009  
≤12 mo prior therapy  .01  B2M ≤2.5 mg/L  .03  
B2M ≤2.5 mg/L  .01  ≤12 mo prior therapy  .4  
Age <65 yr  .2 Age <65 yr  .8 

This report confirms our previous experience that age per se does not affect outcome after autotransplant for MM, whether examined as a continuous or categorical variable. This finding may be due, in large part, to the availability of adequate quantities of CD34+cells in young and old patients, assuring comparable durations of neutropenia and thrombocytopenia and, thus, minimizing the risk of infection and other toxicities. A cumulative injury from a second HDT cycle to either the bone marrow micro-environment or other critical organs was not apparent in either age group. We confirm, in both young and old patients, the importance of cytogenetics and B2M as key variables for sustained disease control. On the basis of our results, there is no biological justification for an age-discriminant policy for MM therapy. In fact, the incidence and nature of complex chromosomal aberrations did not differ among young and old, although different pathogenetic mechanisms may still be revealed at the molecular level.

Palumbo et al recently reported on the safety and efficacy of sequential HDT regimens with cyclophosphamide and melphalan (“CM” regimen)19 as well as repeated sub-myeloablative doses of MEL100 with PBSC support.20 In preparation for regular inclusion of older subjects in HDT trials, we are currently evaluating tandem autotransplants in patients 70 years and older, using MEL140 with the first HDT cycle and, depending on tolerance and antitumor effect, MEL140 or MEL200 with the second transplant. There are no exclusions according to renal function (which we have shown not to affect MEL clearance).21 We conclude that optimal therapy for MM with PBSC-supported high-dose melphalan therapy should not be withheld from the majority of older patients presenting with MM who deserve optimal control of their disease and, thereby, gaining hopefully many years of high-quality life.

The authors greatly acknowledge the dedicated services of the nursing staff, the confidence of many referring physicians who have entrusted us with their patients’ medical care, and to Caran Hammonds for excellent secretarial assistance.

Supported in part by Grant No. CA55819 from the National Cancer Institute, Bethesda, MD.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Barlogie
B
Plasma cell myeloma
Hematology. William’s Hematology
ed 5
Beutler
E
Lichtman
M
Coller
B
Kipps
T
1995
1109
McGraw-Hill
New York, NY
2
Alexanian
R
Dimopoulos
M
The treatment of multiple myeloma.
N Engl J Med
7
1994
484
3
Barlogie
B
Jagannath
S
Tricot
G
Desikan
KR
Fassas
A
Siegel
D
Advances in the treatment of multiple myeloma
Advances in Internal Medicine, vol 43.
Schrier
R
1998
279
Mosby-Year Book
St Louis, MO
4
Attal
M
Harousseau
JL
Stoppa
AM
Sotto
JJ
Fuzibet
JG
Rossi
JF
Casassus
P
Maisonneuve
H
Facon
T
Ifrah
N
Payen
C
Bataille
R
A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma.
N Engl J Med
335
1996
91
5
Barlogie
B
Jagannath
S
Vesole
D
Tricot
G
Naucke
S
Cheson
B
Mattox
S
Bracy
D
Salmon
S
Jacobson
J
Crowley
J
Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma.
Blood
89
1997
789
6
Vesole
DH
Barlogie
B
Jagannath
S
Crowley
J
Cheson
B
Tricot
G
High dose therapy for refractory multiple myeloma: Improved prognosis with better supportive care and double transplants.
Blood
84
1994
950
7
Barlogie
B
Hall
R
Zander
A
Dicke
K
Alexanian
R
High-dose melphalan with autologous bone marrow transplantation for multiple myeloma.
Blood
67
1986
1298
8
Barlogie
B
Alexanian
R
Dicke
KA
Zagars
G
Spitzer
G
Jagannath
S
Horowitz
L
High-dose chemoradiotherapy and autologous bone marrow transplantation for resistant multiple myeloma.
Blood
70
1987
869
9
Vesole
DH
Tricot
G
Jagannath
S
Desikan
KR
Bracy
D
Miller
L
Cheson
B
Crowley
J
Barlogie
B
Autotransplants in multiple myeloma: What have we learned?
Blood
88
1996
838
10
Tricot
G
Sawyer
J
Jagannath
S
Bracy
D
Mattox
S
Vesole
D
Naucke
S
Barlogie
B
Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities.
Blood
86
1995
4250
11
Tricot
G
Sawyer
J
Jagannath
S
Desikan
KR
Siegel
D
Naucke
S
Mattox
S
Bracy
D
Munshi
N
Barlogie
B
The unique role of cytogenetics in the prognosis of patients with myeloma receiving high dose therapy and autotransplants.
J Clin Oncol
15
1997
2659
12
Tricot
G
Jagannath
S
Vesole
DH
Nelson
J
Tindle
S
Miller
L
Cheson
B
Crowley
J
Barlogie
B
Peripheral blood stem cell transplants for multiple myeloma—Identification of favorable variables for rapid engraftment in 225 patients.
Blood
85
1995
588
13
Desikan
KR
Barlogie
B
Jagannath
S
Vesole
DH
Siegel
D
Fassas
A
Munshi
N
Singhal
S
Mehta
J
Tindle
S
Nelson
J
Bracy
D
Mattox
S
Tricot
G
Comparable engraftment kinetics following peripheral blood stem cell infusion mobilized with G-CSF with or without cyclophosphamide in multiple myeloma.
J Clin Oncol
16
1998
1547
14
Smith
AH
Kark
JD
Cassel
JC
Spears
GFS
Analysis of prospective epidemiologic studies by minimum distance case-control matching.
Am J Epidemiol
105
1997
567
15
Barlogie
B
Alexanian
R
Pershouse
M
Smallwood
L
Smith
L
Cytoplasmic immunoglobulin content in multiple myeloma.
J Clin Invest
76
1985
765
16
Kaplan
EL
Meier
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc
53
1958
457
17
Mantel
N
Evaluation of survival data and two new rank order statistics arising in its consideration.
Cancer Chemother Rep
50
1966
163
18
Cox
DR
Regression models and life-tables (with discussion).
J R Stat Soc Series B
34
1972
187
19
Palumbo
A
Pileri
A
Triolo
S
Omede
P
Bruno
B
Cirvegna
G
Galliano
M
Frieri
R
Boccadoro
M
Multicyclic, dose-intensive chemotherapy supported by hemopoietic progenitors in refractory myeloma patients.
Bone Marrow Transplant
19
1997
23
20
Palumbo
A
Pilerio
A
Triolo
S
Bringhen
S
Rus
C
Triolo
R
Ravaglia
R
Boccadoro
M
Intensified therapy with stem cell support (MEL100) for elderly myeloma patients.
Blood
90
1997
231a
(abstr, suppl 1)
21
Tricot
G
Alberts
DS
Johnson
CS
Roe
DJ
Dorr
RT
Vesole
DH
Jagannath
S
Meyers
R
Barlogie
B
Safety of autotransplants with high dose melphalan in renal failure: A pharmacokinetic and toxicity study.
Clin Cancer Res
6
1996
947

Author notes

Address reprint requests to B. Barlogie, MD, PhD, University of Arkansas for Medical Sciences, 4301 W Markham Slot 776, Little Rock, AR 72205.

Sign in via your Institution