Coverage Policy Manual
Policy #: 2000009
Category: Medicine
Initiated: November 1992
Last Review: February 2024
  HDC & Autologous Stem &/or Progenitor Cell Support-Multiple Myeloma

Description:
Multiple myeloma (MM) is a systemic malignancy of plasma cells that represents approximately 10% of all hematologic cancers. It is treatable but rarely curable. At diagnosis, most patients have generalized disease, and the selection of treatment is influenced by patient age, general health, prior therapy, and the presence of disease complications (Kyle, 2008; Palumbo, 2009; Durie, 2006).
 
The disease is staged by estimating tumor mass, based on various clinical parameters such as hemoglobin, serum calcium, number of lytic bone lesions, and the presence or absence of renal failure. Multiple myeloma usually evolves from an asymptomatic premalignant stage (termed monoclonal gammopathy of undetermined significance). Treatment is usually reserved for patients with symptomatic disease (usually progressive myeloma), whereas asymptomatic patients are observed because there is little evidence that early treatment of asymptomatic MM prolongs survival compared with therapy delivered at the time of symptoms or end-organ damage (Kyle, 2008; Palumbo, 2009). In some patients, an intermediate asymptomatic but more advanced premalignant stage is recognized and referred to as smoldering MM. The overall risk of disease progression from smoldering to symptomatic MM is 10% per year for the first 5 years, approximately 3% per year for the next 5 years, and 1% for the next 10 years (Kyle, 2008; Palumbo, 2009).
 
In the prechemotherapy era, the median survival for a patient diagnosed with MM was approximately 7 months. After the introduction of chemotherapy (eg, the alkylating agent melphalan in the 1960s), prognosis improved, with a median survival of 24 to 30 months and 10-year survival of 3%. In a large group of patients with newly diagnosed MM, there was no difference in overall survival reported during a 24-year period from 1971 to 1994, with a trend toward improvement during 1995 to 2000, and a statistically significant benefit in overall survival during 2001 to 2006 (Palumbo, 2009). These data suggested that autologous HCT was responsible for the trends during 1994 to 2000, while novel agents have contributed to the improvement since 2001.
 
The introduction of novel agents and better prognostic indicators has been the major advances in the treatment of this disease (Reece, 2009). Novel agents such as the proteasome inhibitors (eg, bortezomib), the monoclonal antibody daratumumab, and the immunomodulatory derivatives thalidomide and lenalidomide first showed efficacy in relapsed and refractory myeloma and now have been integrated into first-line regimens (Reece, 2009; Qiao, 2015; Rajkumar, 2020). With the introduction of these novel treatments, it is now expected that most patients with MM will respond to initial therapy, and only a small minority will have refractory disease.  
 
High dose chemotherapy (HDC) involves the administration of cytotoxic agents using doses several times greater than the standard therapeutic dose.  In some cases, whole body or localized radiotherapy is also given and is included in the term HDC when applicable.  HDC results in marrow ablation and thus HDC is accompanied by a reinfusion of stem cells in order to repopulate the bone marrow.  
  
Sources of Stem Cells:
Autologous stem cells may be harvested from the patient's bone marrow or peripheral blood.  Peripheral cells are harvested via one or more pheresis procedures.  Pretreatment with chemotherapy and/or hemapoietic growth factors can increase the number of circulating stem cells .Syngeneic stem cells refer to genetically identical bone marrow or peripheral stem cells harvested from an identical twin.    
 
Reimbursement for high dose chemotherapy (HDC) with stem and/or progenitor cell transplant that has been pre-authorized is made as a global fee limited to the lesser of billed charges or the average allowable charge authorized by the Blue Quality Centers for Transplant in the geographic region where the transplant is performed.  This global payment includes all related transplant services including institutional, professional, ancillary, and organ procurement.  The global period begins one day prior to the date of the transplant and continues for 48 days after the transplant.  This covers the inpatient/outpatient stay and provides a per diem outlier payment if necessary.  This global fee also includes the cost of complications arising from the original procedure when services are rendered within the global postoperative period for the particular transplant.
 
Treatment of POEMS syndrome (also known as osteosclerotic myeloma, Crow-Fukase syndrome, or Takasuki syndrome) is addressed in a separate policy # 2013036.
 

Policy/
Coverage:
Effective March 2011
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
High dose chemotherapy with autologous bone marrow, stem cell, or progenitor cell support for the treatment of myeloma meets primary coverage criteria for effectiveness for the treatment of:
    • newly diagnosed or responsive multiple myeloma;
    • responsive myeloma that has relapsed after a durable complete or partial remission following an initial autologous transplant.
 
Tandem high-dose chemotherapy with autologous stem-cell support to treat newly diagnosed or responsive multiple myeloma meets primary coverage criteria for effectiveness and is covered when medically indicated.  Only two (2) courses of therapy given in tandem are covered.
   
Tandem high-dose chemotherapy is considered to be a second course of myeloablative chemotherapy followed by autologous stem-cell support given within  3 to 6 months of the initial course.
  
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
An initial treatment of myeloablative chemotherapy-autologous stem cell transplant followed by non-myeloablative chemotherapy and allogeneic stem-cell support from a HLA-identical sibling donor does not meet member certificate of benefit primary coverage criteria for safety and effectiveness.  
  
Note- For purposes of this policy the term responsive is defined as a tumor showing either a complete or partial remission.  Partial remission is defined as at least a 50% reduction in tumor burden, typically measured in terms of serum levels of beta-2 microglobulin or monoclonal immunoglobulins, both considered tumor markers for multiple myeloma.  
  
Refractory or resistant is defined as a tumor response of less than 50%.
  
High-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is not covered based on benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.  
   
For contracts without primary coverage criteria, high-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is considered investigational and is not covered.  Investigational services are exclusions in the member certificate of coverage.  
 
Effective 2007
High dose chemotherapy with autologous bone marrow, stem cell, or progenitor cell support for the treatment of myeloma meets primary coverage criteria for effectiveness for the treatment of:
    • newly diagnosed or responsive multiple myeloma;
    • responsive myeloma that has relapsed after a durable complete or partial remission following an initial autologous transplant.
 
Tandem high-dose chemotherapy with autologous stem-cell support to treat newly diagnosed or responsive multiple myeloma meets primary coverage criteria for effectiveness and is covered when medically indicated.  Only two (2) courses of therapy given in tandem are covered.
   
Tandem high-dose chemotherapy is considered to be a second course of  myeloablative chemotherapy followed by autologous stem-cell support given within 3 to 6 months of the initial course.
  
An initial treatment of myeloablative chemotherapy-autologous stem cell transplant followed by non-myeloablative chemotherapy and allogeneic stem-cell support from  an HLA-identical sibling donor meets member certificate of benefit primary coverage criteria for newly diagnosed patients 65 years of age or less and in otherwise reasonably good health.
  
For purposes of this policy the term responsive is defined as a tumor showing either a complete or partial remission.  Partial remission is defined as at least a 50% reduction in tumor burden, typically measured in terms of serum levels of beta-2 microglobulin or monoclonal immunoglobulins, both considered tumor markers for multiple myeloma.  
  
Refractory or resistant is defined as a tumor response of less than 50%.
  
High-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is not covered based on benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.  
   
For contracts without primary coverage criteria, high-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is considered investigational and is not covered.  Investigational services are exclusions in the member certificate of coverage.  
 
 
 
Effective prior to 2007
High dose chemotherapy with autologous bone marrow, stem cell, or progenitor cell support for the treatment of myeloma meets primary coverage criteria for effectiveness and is covered:
    • to treat newly diagnosed or responsive multiple myeloma;
    • to treat responsive myeloma that has relapsed after a durable complete or partial remission following an initial autologous transplant.
 
Tandem high-dose chemotherapy with autologous stem-cell support to treat newly diagnosed or responsive multiple myeloma meets primary coverage criteria for effectiveness and is covered when medically indicated.  Only two (2) courses of therapy given in tandem are covered.
 
High-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is not covered based on benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.  
 
For contracts without primary coverage criteria, high-dose chemotherapy with autologous stem-cell support to treat multiple myeloma in a refractory relapse is considered investigational and is not covered.  Investigational services are exclusions in the member certificate of coverage.
 
An initial course of high-dose chemotherapy with autologous stem-cell support followed by non-marrow-ablative chemotherapy and allogeneic stem-cell support (i.e., “mini-transplant”) to treat multiple myeloma is not covered based on benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.  
 
For contracts without primary coverage criteria, an initial course of high-dose chemotherapy with autologous stem-cell support followed by non-marrow-ablative chemotherapy and allogeneic stem-cell support (i.e., “mini-transplant”) to treat multiple myeloma is considered investigational and is not covered. Investigational services are exclusions in the member certificate of coverage.
 
 

Rationale:
Single Transplant
The policy on high-dose chemotherapy with autologous stem cell support (HDC AuSCS) as treatment for multiple myeloma specifically looked at patients with newly diagnosed, responsive multiple myeloma or refractory or resistant myeloma. Responsive myeloma is defined as tumors achieving a complete or partial (at least 50% tumor reduction) response to chemotherapy, while resistant or refractory multiple myeloma is defined as those tumors achieving a less than 50% reduction in tumor burden.   
 
Newly Diagnosed or Responsive Multiple Myeloma
The available data support the conclusion that HDC AuSCS is at least as effective, and may be more effective, than conventional dose chemotherapy for improving the health outcomes of the above patients. In a key randomized trial, the outcomes of high-dose therapy were clearly better than conventional chemotherapy.
  
Resistant or Refractory Multiple Myeloma
In contrast, insufficient data were available to support a conclusion regarding the outcomes HDC AuSCS in patients with refractory myeloma. Most of the data consisted of uncontrolled clinical series of patients. No randomized controlled trials were reported.
  
Tandem Transplant
This policy regarding tandem transplantation references two 1998 Blue Cross Blue Shield Association Technology Evaluation Center assessments, focusing on tandem transplants for newly diagnosed or responsive multiple myeloma and resistant multiple myeloma. The following conclusions were offered:
 
Newly Diagnosed or Responsive Multiple Myeloma
Only 5 published studies were found that provided data on the outcomes of treatment in patients receiving tandem transplant. Although one of the studies is a randomized study, the available data are still preliminary and do not permit
conclusions on survival. The University of Arkansas has reported an extensive non-randomized, single-institution case series of multiple myeloma treated with tandem transplant; however, the data compared results to historical controls
treated with conventional-dose regimens and not to a single cycle of HDC AuSCS, considered the gold standard for comparison.
When using previously published outcomes of patients receiving a single cycle of HDC AuSCS as historical controls, the policy found considerable overlap in the results reported for tandem and single transplant for nearly all of the
outcomes of interest. These data were considered inadequate to permit conclusions regarding the health benefits associated with tandem transplant.  Data on the duration of survival after tandem transplant are scant and nearly all
other outcomes data are only available from single-arm studies with highly selected patients. Thus the comparison of outcomes is subject to a high degree of patient selection bias.
  
Refractory or Resistant Multiple Myeloma
Two reports with a total of 69 patients treated at one institution and a third report with 30 patients provided the only data on the outcomes of tandem transplant for treatment of resistant multiple myeloma. There was no control group for direct comparison of outcomes in the most updated reports on the larger series of highly selected patients. The earlier report from this institution and the third paper included non-randomized control groups, but aggregated outcomes for patients with resistant myeloma and those transplanted as part of first-line therapy. In addition, insufficient detail was provided to determine if the patients given tandem transplant for resistant myeloma were sufficiently comparable to those given either single high-dose chemotherapy or conventional-dose salvage therapy to permit conclusions based on indirect
comparison of outcomes from separate studies. Thus the available data were not sufficient to permit conclusion on the outcomes of tandem transplant.
  
High-dose Chemotherapy and Allogeneic Stem Cell Support
No studies directly comparing the outcomes of high-dose chemotherapy with allogeneic stem cell support (HDC Allo SCS) with either conventional chemotherapy or high-dose chemotherapy with autologous stem cell support have been reported. One retrospective study directly compared the outcomes of allogeneic support with those of autologous support. However this report only provided outcomes that were combined for all myeloma patients, regardless of whether their disease was responsive or refractory to treatment.  Indirect comparisons suggest that allogeneic stem cell support is associated with a 39%–55% 5-year survival, while the comparable figure for autologous stem cell support is 36%–52%.
  
A review of the literature since 1996 does not provide data to alter the above conclusion. In a 1999 review of the data regarding allogeneic stem cell support,  Kyle reported a mortality rate of 25% within 100 days and overall transplant-related mortality of approximately 40%. In addition, relapse of multiple myeloma is common such that few patients are cured.  Candidates for HDC Allo SCS tend to be younger than the average age of patients with multiple myeloma and in better overall condition and thus may have a better prognosis no matter what the treatment. Therefore, randomized trials are required to determine whether any possible benefit associated with HDC Allo SCS is truly related to the therapy rather than the underlying patient selection criteria.
  
High-dose Chemotherapy and Allogeneic Stem Cell Support After a Failed Prior Course of High-dose Chemotherapy and Autologous Stem Cell Support
The policy focused on HDC Allo SCS after a prior failed course of HDC Auto SCS in the treatment of a variety of malignancies, including multiple myeloma.  There were inadequate data to permit conclusions about this treatment strategy.
 
2007 Update
Research continues on many aspects of treatment for multiple myeloma, such as chemotherapy regimens and SCT. The Web site, www.clinicaltrials.gov, lists 305 studies relating to multiple myeloma currently recruiting patients. The current policy may change as additional results, especially with longer follow-up, are released. This review resulted in a policy statement regarding tandem SCT with an autograft followed by an allograft in younger, newly diagnosed patients.  This decision was based primarily on a recent study (Bruno, 2007) using “genetic randomization,” i.e., 80 patients with an HLA-identical sibling were allowed to chose allografts or autografts for the second transplant (58 completed an autograft/allograft sequence) and 82 without an HLA-identical sibling who were assigned to tandem autografts (46 completed the double autograft sequence). The transplantation was preceded by chemotherapy with VAD. The validity of the “genetic randomization” is open to question, but the results among those completing tandem transplantation showed a higher complete response rate at the completion of the second transplant for the autograft/allograft group (55%) than for the autograft/autograft group (26%; p=0.004). Analyzing the group with HLA-identical siblings versus those without, in a pseudo intention-to-treat analysis, event-free survival and OS were significantly longer in the group with HLA-identical siblings. However, it is difficult to gauge the impact of the different percentages in each group who actually complete treatment. The treatment-related mortality rate at 2 years was 2% in the double autograft group and 10% in the autograft/allograft group; 32% of the latter group has extensive, chronic GVHD.
 
A meta-analysis of randomized controlled trials compared chemotherapy versus myeloablative chemotherapy with single auto-SCT (Koreth, 2007).  The 9 trials that met the selection criteria (N=2,411) started enrolling patients in the 1990s and include the 2 studies mentioned above that did not detect a survival benefit from myeloablative chemotherapy (Child, 2003; Attal, 1996). The authors of the meta-analysis concluded that myeloablative therapy with auto-SCT increased the likelihood of progression-free survival (hazard of progression=0.75; 95% CI: 0.59-0.96) but not OS (hazard of death=0.92; 95% CI: 0.74-1.13); the odds ratio for treatment-related mortality was 3.01 (95% CI: 1.64-5.50) in the group with auto-SCT. However, the effects of myeloablative chemotherapy and auto-SCT may have been diluted by the fact that up to 55% of patients in the standard chemotherapy group received myeloablative chemotherapy with auto-SCT as salvage therapy when the multiple myeloma progressed. This could account for the lack of a significant difference in OS between the two groups in the study.
 
Finally, results of the Bologna 96 clinical study have been released (Cavo, 2007), comparing single with double autologous SCT (n=321). Patients undergoing tandem auto-SCT were more likely than those with a single SCT to attain at least a near complete response (47% vs. 33%; p=0.008), to prolong relapse-free survival (median, 42 vs. 24 months; p<0.001), and extend event-free survival (median, 35 vs. 23 months; p=0.001). There was a difference between single and double auto-SCT only for patients who initially responded to VAD. There was no significant difference between the groups in treatment-related mortality (3%–4%). This study supports the use of tandem autografts; it does not address the costs and benefits of novel chemotherapy regimens versus auto-SCT, which is currently being studied, or of autografts versus allografts for the second round of SCT.
 
2008 Update
The policy was updated with a literature search using PubMed in June 2008. Treatment of multiple myeloma has been evolving over the past few years with the use of newer agents including bortezomib, thalidomide, and lenalidomide. A recent review (Koreth, 2007) noted that, “the need for early ASCT [autologous SCT] in an era of new drugs is the most important clinical question in myeloma today.”  
For those with primary progressive disease (disease progression during induction therapy), a risk-adapted approach to treatment is recommended (Stewart, 2007; Fonseca , 2007). This risk-adapted approach is based on several large studies that have shown that patients defined as high risk genetically do not derive durable responses to auto-SCT strategies and may relapse within 1 year of treatment. However, patients with primary progressive disease who do not have these characteristics are considered candidates for auto-SCT. Emerging studies suggest that early introduction of bortezomib results in better quality of response and prolonged survival for high-risk patients.
 
Stewart (2007) suggests an approach to separate the 25% of patients at high risk from those at standard risk. This involves the detection of t(4:14), t(14:16), or 17p deletion by FISH assay, chromosome 13 deletion or hypodiploidy by karyotyping, or plasma cell labeling index greater than 3%. Finding one abnormality identifies a patient at high risk in this approach. In addition, patients with beta-2-microglobulin levels greater than 5.5 mg per liter are often considered high risk.
 
Interest continues in use of allogeneic SCT in multiple myeloma. As noted in a recent review (Bensinger, 2007), despite improvements in survival for multiple myeloma patients, the disease remains incurable for most.  Allogeneic SCT is potentially curative, due in part to a graft-versus-myeloma effect. However, high transplant-related mortality with allo-SCT is a major limitation to wider use of this modality. While mortality can be reduced through the use of RIC regimens, this comes at a cost of higher rates of disease progression and relapse. Studies are ongoing in an attempt to improve overall outcomes of allogeneic SCT. In addition, the risk of chronic GVHD is significant with RIC-allo-SCT.
 
2009 Update
Using search terms myeloma and transplant at www.clinicaltrials.gov results in identification of 555 trials, 251 of which are recruiting.  A PubMed literature search through Sept 2009 was done.
 
Rotta et al (2009) reported long term results of 102 patients treated with auto/allo HCT.  Forty-two percent of the patients developed grade 2 to 4 GVHD and 74% developed chronic GVHD.  Among 95 patients with detectable disease, 59 achieved complete remissions. Five-year nonrelapse mortality was 18% with 95% of that attributable to GVHD or infection.
 
Bensinger, in another review article in 2009, continues to state that the disease remains incurable for all but a small fraction of patients despite good response rates with new drugs and the mortality associated with allogeneic transplants.
 
Vesole (2009) reported results of 32 patients enrolled in a Phase II trial of autologous stem cell transplant followed by a mini-allogeneic stem cell transplant.  Twenty-three patients completed both transplants.  There were 7 complete and 11 partial remissions, 2 with no response and 3 were not evaluable.  Acute grade III-IV GVHD was seen in 4 patients, chronic GVHD in 13 patients.  “Because a plateau in PFS or OS was not observed with this treatment approach even in patients achieving CR, we suggest that future studies use posttransplantation maintenance therapy.”
 
In a review and meta-analysis of tandem versus single autologous hematopoietic cell transplantation to treat multiple myeloma, Kumar (2009) looked at six RCTs enrolling 1803 patients meeting inclusion criteria.  Patients with tandem AHCT did not have better OS or EFS.  They did have statistically significant response rates but this was associated with a statistically significant increase in treatment-related mortality.
No literature was identified that would support a revision in the current coverage statements.
 
2011 Update
In December 2010, at the American Society of Hematology annual meeting in Orlando, Drs. Amrita Krishnan and Edward Stadtmauer presented 3-year preliminary results of a phase III trial that enrolled 710 patients with standard risk myeloma in 43 centers in the United States.  All patients received high-dose melphalan and an autologous stem cell transplant.  The 484 patients without a sibling match were to receive a second autologous transplant with Melphalan ("allo-allo").  The 226 who had a sibling match were to received a nonmyeloablative conditioning regimen followed by an allogeneic stem cell transplant and 2 Gy of total body irradiation ("allo-auto").  In each group 82% went on to a second transplant.  For those patients who received a second transplant progression free survival rates, overall survival rates and progression/relapse rates were similar in both groups.  Treatment related mortality was significantly different in the two groups; 4% for the allo-allo group and 12% for the allo-auto group.  Dr. Stadtmauer commented:  "This is a preliminary result but it certainly is not supportive of routine use of nonmyeloablative allogeneic transplant in standard-risk patients. …With longer-term follow-up, if these results hold out, then I think our enthusiasm will disappear."
 
2013 Update
A search of the MEDLINE database was conducted through September 2013. There was no new information identified that would prompt a change in the coverage statement.
 
2014 Update
A literature search conducted through September 2014 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The EBMT reported an analysis of 413 MM patients who received a related or unrelated RIC allogeneic HSCT for the treatment of relapse or disease progression after a prior autologous HSCT (Auner, 2013). Median age at RIC allogeneic HSCT was 54 years, and 45% of patients had undergone two or more prior autologous transplants. The median OS and PFS from the time of allogeneic transplantation for the entire population were about 25 and 10 months, respectively. Cumulative non-relapse mortality (NRM) at 1 year was about 22%. In a multivariate analysis, cytomegalovirus (CMV) seronegativity of both patient and donor was associated with significantly better PFS, OS and NRM. Patient-donor gender mismatch was associated with better PFS, fewer than two prior autologous transplants was associated with better OS, and shorter time from the first autologous HSCT to the RIC allogeneic HSCT was associated with lower NRM. These results suggest patient and donor CMV seronegativity represent key prognostic factors for outcome after RIC allogeneic HSCT for MM that relapses or progresses following one or more autologous transplants.
 
At 96 months in the EBMT trial, progression-free survival (PFS) and overall survival (OS) were 22% and 49% versus 12% (P = .027) and 36% (P = .030) with autologous/RIC-allogeneic and autologous HSCT, Respectively (Gahrton, 2013). The corresponding relapse/progression rate (RL) was 60% versus 82% (P = .0002). Nonrelapse mortality at 36 months was 13% versus 3% (P = .0004). In patients with the del(13) abnormality corresponding PFS and OS were 21% and 47% versus 5% (P = .026), and 31% (P = .154) Long-term outcome in patients with multiple myeloma was better with autologous/RIC-allogeneic HSCT as compared with autologous only and the autologous/RIC-allogeneic approach seemed to overcome the poor prognostic impact of del (13) observed after autologous transplantation.
 
The role of allogeneic HSCT remains controversial, in particular because of conflicting data from cooperative group trials, but also because of improvement in outcomes that have been observed with proteasome inhibitors, new immune modulatory agents, and the use of post-transplant maintenance therapy. These issues have recently been reviewed and summarized (Giralt, 2014; Giralt, 2014).
 
A single-center series published in 2012 from Mayo Clinic reported a 5-year OS of 94% and a PFS of 75% among 59 patients entered between 1999 and late 2011. A second recent series included 9 advanced POEMS syndrome patients, who had an Eastern Cooperative Oncology Group performance status score of 3 or 4, and were treated with high-dose melphalan therapy followed by autologous stem cell transplantation from 2004 to 2011 (Jang, 2014). Eight patients achieved an initial hematologic response, 4 of whom had complete responses. At a median follow-up of 44 months (range, 8-94 months), 7 patients were alive, with 3-year overall survival rate of 78%. There were no hematologic relapses in the survivors. One patient died of disease progression; the other died of pneumonia despite a hematologic response 3 months after autologous stem cell transplantation. All survivors achieved improvement in general performance status and in clinical response. The responses observed in these patients with advanced POEMS suggest it is a valid treatment option for such cases.
 
2015 Update
A literature search conducted through September 2015 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
In a multicenter, randomized, open-label, phase 3 study from 51 centers across the United Kingdom, between April 16, 2008, and November 19, 2012, Cook and colleagues recruited patients aged at least 18 years with MM who needed treatment for first progressive or relapsed disease at least 18 months after a previous autologous HSCT (NCT00747877) and EudraCT (2006-005890-24) (Cook, 2014). Before randomization, eligible patients received bortezomib, doxorubicin, and dexamethasone (PAD) induction therapy and then underwent peripheral blood stem cell mobilization and harvesting, if applicable. Eligible patients were randomly assigned (1:1) to receive either high-dose melphalan 200 mg/m2 plus salvage autologous HSCT or oral cyclophosphamide (400 mg/m2/wk for 12 weeks). The primary end point was time to disease progression, analyzed by intention to treat. A total of 297 patients were enrolled, of whom 293 received PAD re-induction therapy. Among the latter, 174 patients with sufficient harvest of peripheral blood stem cells were randomly allocated to undergo salvage HSCT (n=89) or receive cyclophosphamide (n=85). After a median follow-up of 31 months, median time to progression was significantly longer in the salvage HSCT group than in the cyclophosphamide group (19 months [95% CI, 16 to 25] vs 11 months [95% CI, 9 to 12]; hazard ratio, 0.36 [95% CI, 0.25 to 0.53]; p<0.001). Frequently reported (>10% of patients) grade 3-4 morbidity with PAD induction, salvage HSCT, and cyclophosphamide were: neutropenia (125 [43%] of 293 patients after PAD and 63 [76%] of 83 patients in the salvage HSCT group vs 11 [13%] of 84 patients in the cyclophosphamide group), thrombocytopenia (150 [51%] after PAD, 60 [72%] vs 4 [5%], respectively), and peripheral neuropathy (35 [12%] after PAD, and none vs none, respectively). This study provides additional evidence for a net benefit of high-dose melphalan plus salvage HSCT when compared with cyclophosphamide in patients with relapsed MM eligible for intensive therapy.
 
Multiple Myeloma
The evidence for autologous hematopoietic stem cell transplantation (HSCT) for upfront treatment in patients who have newly diagnosed multiple myeloma consists of several prospective, randomized controlled trials (RCTs) that compared conventional chemotherapy with high-dose chemotherapy with autologous HSCT. Clinical outcomes of interest include overall survival (OS) and treatment-associated morbidity. In general, the evidence suggests OS rates are improved with autologous HSCT compared with conventional chemotherapy in this setting. Limitations of the published evidence include patient heterogeneity; variability in treatment protocols; short follow-up periods; inconsistency in reporting important health outcomes; and, inconsistency in reporting or collecting outcomes. These limitations notwithstanding, the body of evidence on autologous HSCT for treatment of patients with newly diagnosed multiple myeloma is sufficient to demonstrate improvements in health outcomes
 
The evidence for autologous HSCT for treatment of relapsed MM following autologous HSCT, or refractory disease consists of 1 RCT and a systematic review that summarized data from 4 clinical series of patients who relapsed after a first autologous HSCT. Clinical outcomes of interest include OS and treatment-related morbidity. In general, the evidence suggests OS rates are improved with autologous HSCT compared with conventional chemotherapy in this setting. Limitations of the published evidence include patient heterogeneity; variability in treatment protocols; short follow-up periods; inconsistency in reporting important health outcomes; and, inconsistency in reporting or collecting outcomes. These limitations notwithstanding, the body of evidence on autologous HSCT for treatment of patients with relapsed MM or refractory disease is sufficient to demonstrate improvements in health outcomes.
 
The evidence for tandem autologous HSCT in patients who have MM who fail to achieve at least a near complete or very good partial response after the first transplant in the tandem sequence (ie, refractory disease) consists of 3 RCTs. Clinical outcomes of interest include OS and treatment-related morbidity. The evidence shows tandem autologous HSCT improves OS rates in this setting. Limitations of the published evidence include patient heterogeneity; variability in treatment protocols; short follow-up periods; inconsistency in reporting important health outcomes; and, inconsistency in reporting or collecting outcomes. These limitations notwithstanding, the body of evidence on tandem autologous HSCT for treatment of patients with MM in this setting is sufficient to demonstrate improvements in health outcomes.
 
The evidence for tandem autologous HSCT followed by reduced-intensity conditioning (RIC) allogeneic HSCT in patients who have newly diagnosed MM comprises several RCTs comparing RIC-allogeneic HSCT following a first autologous HSCT with autologous transplants, single or in tandem. These studies were based on “genetic randomization,” that is, patients with an HLA-identical sibling were offered an RIC-allogeneic HSCT following the autologous HSCT, whereas the other patients underwent either 1 or 2 autologous transplants. Clinical outcomes of interest include OS and treatment-related morbidity. Although the body of evidence shows inconsistencies in terms of OS and DFS rates, some studies have shown a survival benefit with tandem autologous-RIC allogeneic HSCT, although at a cost of higher transplant-related mortality compared with conventional treatments. Factors across studies that may account for differing trial results include different study designs; nonuniform preparative regimens; different patient characteristics (including risk stratification); and, criteria for advancing to a second transplant. These limitations notwithstanding, the body of evidence on tandem autologous HSCT followed by RIC allogeneic HSCT for treatment of patients with MM is sufficient to demonstrate improvements in health outcomes.
 
American Society for Blood and Marrow Transplantation (Shah, 2015).
In 2015, the American Society for Blood and Marrow Transplantation (ASBMT) published evidence-based guidelines for the use of HSCT in patients with MM. These guidelines are generally consistent with the conclusions of this evidence review based on published literature through December 31, 2014. ASBMT recognizes that much of the RCT evidence summarized in the 2015 guidelines comes from trials that predate the advent of novel triple therapy induction regimens. Furthermore, advances in supportive care and earlier disease detection has increasingly influenced decision making and allows individual tailoring of therapy. ASBMT guidelines do not address POEMS or other plasma cell dyscrasias besides MM.
 
2017 Update
A literature search conducted through September 2017 did not reveal any new information that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
Randomized Controlled Trials
One 2015 randomized controlled trial (RCT) compared autologous HCT to standard chemotherapy plus lenalidomide, a newer agent for treatment of MM (Gay, 2015). The open-label RCT from 59 centers in Europe and Australia used a 2×2 factorial design to compare 4 groups (1) standard consolidation therapy plus HCT, followed by maintenance with lenalidomide alone, (2) standard consolidation therapy plus HCT, followed by maintenance with lenalidomide and prednisone, (3) consolidation with chemotherapy plus lenalidomide, followed by maintenance with lenalidomide alone, and (4) consolidation with chemotherapy plus lenalidomide, followed by maintenance with lenalidomide plus prednisone. The primary outcome was progression-free survival (PFS). Mean follow-up at the time of publication was 52 months. Median PFS was superior for the HCT group plus standard consolidation (43.3 months; 95% confidence interval [CI], 33.2 to 52.2 months) compared to chemotherapy plus lenalidomide (28.6 months; 95% CI, 20.6 to 36.7 months; p<0.0001). The rate of grade 3 or 4 adverse events was higher for the HCT group than for the chemotherapy groups (hematologic events, 84% vs 26%; gastrointestinal complications, 20% vs 5%; infections, 19% vs 5%; all respectively).
 
Final survival data for the Myeloma X Relapse trial were reported in 2016 (Cook, 2016). The HCT group had superior median OS (67 months; 95% CI, 55 months to not estimable) compared to the chemotherapy group (52 months; 95% CI, 42 to 60 months; p<0.001). Time to disease progression continued to favor the HCT group at the longer follow-up (19 months [95% CI, 16 to 26 months] vs 11 months [95% CI, 9 to 12 months]; p=0.02). There were no further adverse events related to the HCT procedure reported during longer follow-up. The cumulative incidence of second malignancies was 5.2% (95% CI, 2.1% to 8.2%).
 
2018 Update
A literature search conducted using the MEDLINE database through May 2018 did not reveal any new information that would prompt a change in the coverage statement.
 
NEWLY DIAGNOSED MM
 
Early vs Delayed HCT
A 2017 retrospective analysis by Dunavin et al compared survival and relapse rates in 167 patients who were treated for MM between 2002 and 2009 with induction therapy and autologous HCT (Dunavin, 2017). In the first group (n=102), autologous HCT was given no more than 12 months after diagnosis; in the second, autologous HCT was given 12 months or more after diagnosis, although individual reasons for later procedures were not specified. Following a standard induction therapy and preceding transplantation, more patients in the early group had achieved a complete response (CR) or very good partial response than in the late autologous HCT group (46% vs 62%, p=0.036). This difference remained significant after transplantation with patients who were upgraded to very good partial response or CR (early autologous HCT, 77% vs late autologous HCT, 56%; p<0.007). No significant differences were observed between groups for progression-free survival (PFS) or overall survival (OS), which were assessed at 1, 3, and 5 years; however, a difference of 10 months between groups in median PFS was noted (28 months for early autologous HCT patients vs 18 months for late autologous HCT patients). Relapse occurred in 40% of patients in the early autologous HCT group, and 55% of the late autologous HCT group (p=0.55). A variable that did have a significant bearing on PFS between groups was that of risk, with high-risk patients in the early autologous HCT group achieving a median PFS of 25 months, compared with the 11 months achieved by their counterparts in the late autologous HCT group. The results of this study seemed to confirm the observation made by previous studies that patients who achieve a CR are more likely to remain progression-free for significantly longer than those whose response to induction therapy is not very good. Data were lacking on the reason for delayed autologous HCT; another limitation was that patients who received maintenance therapy were excluded from the study.
 
RELAPSED OR REFRACTORY MM
 
Salvage Autologous HCT for Relapsed MM
A 2013 multicenter retrospective study by Michaelis et al evaluated 187 patients drawn from the Center for International Blood and Marrow Transplantation who were treated with a second autologous HCT following relapse or progression of MM (Michaelis, 2013). All but 12% of patients received a second autologous HCT12 months or more after the initial transplantation; prior to a second autologous HCT, only 40% (n=74) of patients were in complete or partial response. In patients whose time from the first transplant to first relapse was greater than 36 months, investigators noted a decrease in the risk of relapse after a second autologous HCT(relative risk, 0.63; 95% CI, 0.49 to 0.97), and an increase in PFS and OS. For such individuals, the 3-year PFS rate was twice that of the cohort at large (26% vs 13%), and 5-year PFS rate (13%) was considerably superior to that of the larger group (5%). A comparison of OS rates showed a similar improvement: while the 5-year OS rate of 29% for the entire cohort was comparable to other studies of a second autologous HCT in relapsed MM, the 5-year OS rate for individuals with a time-to-relapse of 36 months or greater was considerably improved (48%; p=0.026). After 3 years, only 4% (95% CI, 2% to 8%) of patients experienced NRM; however, relapse or disease progression was observed in 82% of patients after 3 years (vs 68% of patients with time-to-relapse 36 months after initial transplant). The investigators acknowledged a lack of data on maintenance regimens, cytogenetics, or staging of individual disease; they also noted that, during the observed time frame (1995-2008), several newer therapies were introduced, which were not accounted for during analysis. However, given findings similar to other retrospective studies during the same period, the investigators concluded that a second autologous HCT is an appropriate salvage therapy for eligible patients.
 
A 2017 review by Ziogas et al included studies of autologous HCT as salvage therapy in patients whose MM has relapsed following an initial autologous HCT (either single or tandem) (Ziogas, 2017). The primary aim of the review was to summarize the circumstances in which a second autologous HCT should be administered, especially as more regimens show potential as salvage or reinduction therapy, including anti-CD38 antibodies, next-generation proteasome inhibitors, or immunomodulatory drugs. The authors noted that most studies have been retrospective, or of small patient samples; however, in 15 of the included studies, more than 40 patients were evaluated. Overall response rates ranged from 55.3% to 97.4%; following a salvage transplant, median PFS across studies varied considerably (range, 8.5-40 months). The questions examined in the review concerned the safety and efficacy of a second autologous HCT, predictors of outcome and best maintenance approach following salvage autologous HCT, and the future of the treatment. Based on general agreement from studies that showed the particular benefit of salvage autologous HCT in patients with longer intervals from the first transplant to initial relapse, reviewers recommended that the treatment is administered to patients with remission of greater than 18 months following initial autologous HCT. Given heterogeneity across studies of novel maintenance therapies, reviewers called for more prospective studies, noting melphalan as a well-established basis for treatment.
 
Tandem Autologous HCT for Relapse After First Autologous HCT
A 2016 review by McCarthy and Holstein summarized current treatment regimens for patients with myeloma who are eligible for autologous HCT or allo-HCT (McCarthy, 2016). Following discussion of studies on induction, salvage, consolidation, and maintenance therapies, reviewers offered recommendations based on the available evidence. Based on 4 studies comparing autologous HCT with chemotherapy alone, reviewers recommended autologous HCT as standard of care for patients who are eligible; additionally, they recommended autologous HCT for the first relapse, based on the pooled hazard ratio of 2 studies showing a benefit in patients given autologous HCT following relapse (hazard ratio, 0.57; p=0.037). Reviewers noted the increasing uncertainty regarding the efficacy and safety of allo-HCT compared with novel therapies; studies directly comparing allo-HCT with autologous HCT lack consistent results. However, RIC allo-HCT has been shown to have some benefit for patients whose disease is high-risk, especially in younger populations. As maintenance therapy, reviewers considered a number of studies evaluating thalidomide (n=8), which had conflicting results, as well as 3 randomized studies of lenalidomide, concluding that the latter treatment is standard of care.
 
2019 Update
A literature search was conducted through May 2019.  There was no new information identified that would prompt a change in the coverage statement.  
 
2020 Update
A literature search was conducted through May 2020.  There was no new information identified that would prompt a change in the coverage statement.  
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The approach to the treatment of newly diagnosed MM (symptomatic) is dictated by eligibility for autologous hematopoietic cell transplantation (HCT) and risk stratification (Rajkumar, 2020). Risk stratification, using fluorescent in situ hybridization and conventional karyotyping divides patients into high- or standard-risk categories.
 
High-risk patients, which comprise approximately 25% of patients with MM, are defined by any of the following cytogenetic findings: a 17p deletion; translocations of chromosomes 4 and 14, chromosomes 14 and 16, chromosomes 14 and 20; or a 1q gain (Rajkumar, 2020). Standard-risk patients are those with hyperdiploidy (translocations of chromosomes 11 and 14 and chromosomes 6 and 14).
 
High-risk patients are generally treated with a bortezomib-based induction followed by autologous HCT and then bortezomib-based maintenance (Rajkumar, 2020)).https://www.evidencepositioningsystem.com/_w_018956f4988fa4d5b181dc7f9051210ca171476bbeea0421/bcbsa_html/BCBSA/html/_blank Standard-risk patients are typically treated with bortezomib-based induction therapy followed by autologous HCT and then maintenance with lenalidomide; however, if the patient is tolerating the induction regimen well, an alternative strategy would be to continue the initial therapy after hematopoietic cell collection, reserving the transplant for the first relapse.
 
Several RCTs have compared autologous HCT with treatment regimens that utilize newer MM agents. Cavo et al conducted a multicenter, randomized, open-label phase 3 study comparing standard-dose intensification therapy with bortezomib, melphalan, and prednisone (n=495) to high-dose melphalan plus autologous HCT (n=702) in patients with newly diagnosed MM (up to 65 years of age) (Cavo, 2020). Within the autologous HCT group, 492 received a single autologous HCT and 210 received a double autologous HCT. Median progression-free survival (PFS) was 56.7 months (95% confidence interval [CI], 49.3 to 64.5) for patients receiving autologous HCT versus 41.9 months (95% CI, 37.5 to 46.9) for those assigned to standard-dose intensification therapy (hazard ratio, 0.73; 95% CI, 0.62 to 0.85; p=0.0001). The 5-year OS rate was 75.1% (95% CI, 71.7 to 78.5) for patients in the autologous HCT group and 71.6% (95% CI, 67.4 to 76.1) for those in the standard-dose intensification therapy group (hazard ratio, 0.90; 95% CI, 0.71 to 1.13; p=0.35). Among patients with high-risk cytogenetic profiles, OS was significantly better with autologous HCT.
 
Attal et al conducted a randomized, open-label phase 3 trial in patients less than 65 years of age with newly diagnosed MM (Attal, 2017). Patients were randomly assigned to receive consolidation therapy with 5 cycles of bortezomib, lenalidomide, and dexamethasone (n=350) or high-dose melphalan followed by autologous HCT and 2 cycles of bortezomib, lenalidomide, and dexamethasone (n=350). With a median follow-up of 43 months, median PFS was 36 months in the non-transplant group versus 50 months in the transplant group (hazard ratio, 0.65; p<0.001). The 4-year PFS rates were 35% and 50% for non-transplant and transplant groups respectively (p<0.001), while 4-year OS rates were 82% and 81%, respectively (p=0.43). Median OS was not reached for either group.
 
In the RCT by Cavo et al described in the section above, patients who were assigned to receive autologous HCT at a center that performed double autologous HCT were randomly assigned to receive either single (n=209) or double (n=210) autologous HCT (Cavo, 2020). Outcomes were compared between these subgroups in a secondary analysis. Double autologous HCT significantly improved rates of 5-year PFS (53.5% vs. 44.9%; hazard ratio, 0.74; 95% CI, 0.56 to 0.98; p=0.036) and 5-year OS (80.3% vs. 72.6%; hazard ratio, 0.62; 95% CI, 0.41 to 0.93; p=0.022) compared to single autologous HCT. Patients with high-risk cytogenetic profiles appeared to attain a greater magnitude of benefit with double HCT versus single HCT, compared to patients with standard-risk profiles.
 
A systematic review and meta-analysis by Mian et al specifically sought to examine the impact of autologous HCT in patients aged 65 years or older with newly-diagnosed MM (Mian, 2020). This review included data from 2 RCTs and 6 observational studies. In a pooled analysis of the observational studies, autologous HCT was associated with favorable effects on OS compared to non-HCT therapy (hazard ratio, 0.44; 95% CI, 0.34 to 0.58; p<0.0001). However, in the pooled analysis of RCT data, the impact of autologous HCT on OS was uncertain (hazard ratio, 0.94; 95% CI, 0.25 to 3.54, p=0.93). Observational data also showed higher CR rates with autologous HCT (odds ratio, 5.06; 95% CI, 2.60 to 9.88; p<0.0001). The authors of the review concluded that autologous HCT may improve the OS and CR rates in elderly patients based on observational data, but the quality of the evidence is very low and more studies are needed.
 
Stadtmauer et al reported a randomized phase 3 study in patients with symptomatic MM who received at least 2 cycles of any regimen as initial systemic therapy without disease progression and who were within 2 to 12 months of the first dose of initial therapy (Stadtmauer, 2019). Patients were randomly assigned to 1 of 3 treatment arms: autologous HCT (n=257), tandem autologous HCT (n=247), or autologous HCT plus 4 cycles of lenalidomide, bortezomib, and dexamethasone (n=254). Rates of 38-month PFS were similar across groups (58.5%, 57.8%, and 53.9% for tandem HCT, autologous HCT plus lenalidomide/bortezomib/dexamethasone, and autologous HCT respectively), as were rates of 38-month OS (81.8%, 85.4%, and 83.7%, respectively). However, 32% of patients in the tandem group did not receive the second HCT. Results of this study differed from those of the Cavo et al study described above. This may be related to differences in initial therapy; in the Cavo et al study, patients received a prespecified number of induction therapy cycles that did not include immunomodulatory agents (eg, lenalidomide), while the majority of patients in this study received immunomodulatory agents as part of their initial therapy prior to transplant. Additionally, more patients in the Cavo et al study underwent tandem HCT as assigned (only 20% did not receive the second transplant) (Cavo, 2020).
 
Goldschmidt et al conducted a randomized, open-label, multicenter phase 3 study (the ReLApsE trial) in patients aged 18 to 75 years with a first to third relapse of MM (Goldschmidt, 2020). These patients had previously undergone autologous HCT and attained remission of at least 12 months prior to relapse. Patients were randomized to receive a repeat autologous HCT (n=139) or continuous therapy with lenalidomide plus dexamethasone (n=138). Patients who underwent repeat autologous HCT also received reinduction therapy with lenalidomide plus dexamethasone, salvage high-dose chemotherapy with melphalan, and lenalidomide maintenance. In the primary ITT analysis, no significant differences were seen in PFS (median, 20.7 months vs. 18.8 months for transplant vs. control; hazard ratio, 0.87; 95% CI, 0.65 to 1.16; p=0.34) or OS (median not reached in the transplant group vs. 62.7 months in the control arm; hazard ratio, 0.81; 95% CI, 0.52 to 1.28; p= 0.37). However, only 71% of patients assigned to the transplant group actually underwent salvage high-dose chemotherapy and autologous HCT. Post hoc analyses found that the patients who received salvage high-dose chemotherapy and autologous HCT had a trend toward superior PFS compared to the control group, and statistically superior OS (median not reached vs. 57 months; hazard ratio, 0.56; 95% CI, 0.32 to 0.99; p=0.046).
 
A retrospective study by Ikeda et al examined outcomes of a second HCT (either allo-HCT [n=192] or repeat autologous HCT [n=334]) in patients with relapsed or progressive MM after a first autologous HCT (Ikeda, 2019). Rates of 5-year OS were 23.8% after allo-HCT and 33.7% after repeat autologous HCT; however, differences in these rates were likely influenced by differences in baseline characteristics, such as age, performance status, time from initial HCT, and response to chemotherapy before HCT. Patients were assigned risk categories based on response to reinduction, performance status, and time from initial HCT; in intermediate-risk patients (the largest risk subgroup), OS rates were higher with repeat autologous HCT versus allo-HCT (28.2% vs. 21.5%; p<0.004). No significant differences were noted in the low- and high-risk subgroups.
 
In 2019, the American Society of Clinical Oncology (ASCO) published practice guidelines for the treatment of multiple myeloma (MM) (Mikhael, 2019). The guidelines recommend offering up-front transplant to all eligible patients, although delayed HCT may be considered in select patients. Salvage or delayed HCT may be used as consolidation at first relapse in patients who choose not to proceed with HCT initially. Tandem autologous HCT and allogeneic HCT (allo-HCT) should not be routinely recommended. However, up-front tandem autologous HCT can be considered for select high-risk patients or those with a suboptimal response to the initial transplant; allo-HCT may be considered in select high-risk patients in the context of a clinical trial. For relapsed MM, autologous HCT, if not received after primary induction therapy, should be offered to transplant-eligible patients. Repeat HCT may be considered in relapsed MM if progression-free survival after the first transplant was 18 months or greater.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2022. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The results of a 5-year follow-up of the trial by Stadtmauer et al were posted to Clinicaltrials.gov (NCT02322320) but have not been identified in a peer-reviewed journal (Clinical Trials, 2020). Per Clinicaltrials.gov, the proportion of patients achieving PFS was similar for autologous HCT (45%), tandem autologous HCT (47.7%), and autologous HCT plus lenalidomide, bortezomib, and dexamethasone (44.1%); pairwise comparisons between treatment arms did not reach statistical significance. Likewise, the proportion of patients achieving OS was similar for autologous HCT (76.4%), tandem autologous HCT (74.7%), and autologous HCT plus lenalidomide, bortezomib, and dexamethasone (75.4%); pairwise comparisons between treatment arms did not reach statistical significance
 
The National Comprehensive Cancer Network (NCCN) guideline for multiple myeloma (v3.2022) states that autologous HCT is the preferred option after induction therapy in transplant-eligible patients, but a delayed HCT after early stem cell collection and storage is appropriate as well (category 1 recommendation) (NCCN, 2021). A repeat HCT can be considered for refractory/progressive disease after primary treatment in patients with prolonged response to initial HCT.
 
The NCCN guideline for multiple myeloma (v3.2022) recommends collecting enough stem cells for 2 transplants in younger patients if tandem transplant or salvage transplant would be considered (NCCN, 2021). A tandem transplant with or without maintenance therapy can be considered for all patients who are candidates for HCT and is an option for patients who do not achieve at least a very good partial response after the first autologous HCT and those with high-risk features.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Several RCTs have compared autologous HCT with treatment regimens that utilize newer MM agents. Richardson et al conducted a US-based, multicenter, open-label RCT comparing lenalidomide, bortezomib, and dexamethasone alone with the lenalidomide, bortezomib, and dexamethasone regimen in addition to autologous HCT plus melphalan in patients with newly diagnosed multiple myeloma (Richardson, 2022). All patients received daily maintenance lenalidomide until disease progression, unacceptable toxicity, or withdrawal from treatment or the trial. Patients treated with chemotherapy alone (n=357) had lower median PFS (46.2 months) compared with those who received chemotherapy and autologous HCT (n=365; 67.5 months). Patients who received chemotherapy only had higher rates of disease progression or death at a median follow-up of 76 months (HR, 1.53; 95% CI, 1.23 to 1.91; p<.001). Overall survival was similar between groups. Grade 3 or higher treatment-related adverse events were higher in patients undergoing HCT (94.2% vs. 78.2%).
 
Villalba et al analyzed data from 35 hospitals in the Spanish Myeloma Group (Villalba, 2022). Patients (N=213) with newly diagnosed multiple myeloma and high-risk cytogenetics underwent single (n=142) or tandem (n=71) autologous HCT. At a median follow-up of 31 months, PFS was nonsignificantly longer with tandem HCT compared with single HCT (48 vs. 41 months; p=.33). Patients receiving tandem HCT were younger, had more advanced stage disease, and a higher plasma cell infiltration at diagnosis. More patients in the singe-transplant group died by the time of analysis than those undergoing tandem transplant although this was not statistically significant (23% vs. 12.7%; p=.09). The authors concluded that tandem HCT partly overcomes the poor prognosis of high-risk cytogenetics when compared with a single HCT but noted further study is needed.
 
2024 Update
Annual policy review completed with a literature search using the MEDLINE database through January  2024. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The ASTCT updated guidance for transplantation and cellular therapies in MM in 2022 (Dhakal, 2022). The panel endorsed continued use of autologous HCT for patients with newly diagnosed MM as a standard-of-care option and did not recommend front-line use of allo-HCT and CAR-T outside the setting of a clinical trial. For patients not undergoing autologous HCT upfront, the panel recommended its use in first relapse. The panel also encouraged allo-HCT in relapsed/refractory MM setting only in the context of clinical trial.
 
In 2020, the ASTCT published a guideline on indications for HCT and immune effector cell therapy (Kanate, 2020). Regarding plasma cell dyscrasias, the guideline states that MM remains the most common indication for autologous HCT. For rarer plasma cell dyscrasias like POEMS syndrome, autologous HCT may be considered a clinical option on the basis of single-center and registry data.

References: 1996 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 14.

1996 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 28.

1998 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 26.

1998 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 8.

Alegre A, Díaz-Mediavilla J, San-Miguel J, et al.(1998) Autologous peripheral blood stem cell transplantation for multiple myeloma: a report of 259 cases from the Spanish Registry. BMT 1998; 21:133-40.

Alexanian R, Dimopoulos M, Smith T, et al.(1994) Limited value of myeloablative therapy for late multiple myeloma. Blood 1994; 83:512-6.

Alexanian R, Dimopoulos M.(1994) The treatment of multiple myeloma. NEJM 1994; 330:484-9.

Alexanian R, Dimopoulos MA, Hester J, et al.(1994) Early myeloablative therapy for multiple myeloma. Blood 1994; 84:4278-82.

Anderson KC, Andersen J, Soiffer R, et al.(1993) Monoclonal antibody-purged autologous bone marrow transplantation therapy for multiple myeloma. Blood 1993; 82:2568-76.

Armitage JO.(1994) Bone marrow transplantation. NEJM 1994; 330:827-38.

Attal M, Harousseau JL, Stoppa AM, et al.(1996) A prospective randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. NEJM 1996; 335:91-97.

Attal M, Lauwers-Cances V, Hulin C, et al.(2017) Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N Engl J Med. Apr 06 2017; 376(14): 1311-1320. PMID 28379796

Auner HW, Szydlo R, van Biezen A, et al.(2013) Reduced intensity-conditioned allogeneic stem cell transplantation for multiple myeloma relapsing or progressing after autologous transplantation: a study by the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. Nov 2013;48(11):1395-1400. PMID 23708704

Barlogie B, Jagannath S, Vesole D, et al.(1995) Autologous and allogeneic transplants for multiple myeloma. Semin Hematol 1995; 32:31-44.

Barlogie B, Jagannath S, Vesole D, et al.(1997) Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood 1997; 89:789-793.

Baron F, Storb R, et al.(2006) Factors associated with outcomes in allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning after failed myeloablative hematopoietic cell transplantation. J Clin Oncol, 2006; 24(25):4150-7.

Bensinger WI, Rowley SD, Demirer T, et al.(1996) High-dose therapy followed by autologous hematopoietic stem-cell infusion for patients with multiple myeloma. J Clin Oncol 1996; 14:1447-56.

Bensinger WI.(2009) Role of autologous and allogeneic stem cell transplantation in myeloma. Leukemia, 2009; 23:442-8.

Bjorkstrand B, Goldstone AH, Ljungman P, et al.(1994) Prognostic factors in autologous stem cell transplantation for multiple myeloma: an EBMT Registry Study. Leuk Lymphoma 1994; 15:265-72.

Bjorkstrand B, Ljungman P, Bird JM, et al.(1995) Double high-dose chemoradiotherapy with autologous stem cell transplantation can induce molecular remissions in multiple myeloma. BMT 1995; 15:367-71.

Bladé J, López-Guillermo A, Bosch F, et al.(1994) Impact of response to treatment on survival in multiple myeloma: results in a series of 243 patients. Br J Haematol 1994; 88:117-21.

Bladé J, San Miguel J, Sanz-Sanz MA, et al.(1992) Treatment of melphalan-resistant multiple myeloma with vincristine, BCNU, doxorubicin, and high-dose dexamethasone (VBAD). Eur J Cancer 1992; 29A:57-60.

Browman GP, Belch A, Skillings J, et al.(1992) Modified Adriamycin-vincristine-dexamethasone (m-VAD) in primary refractory and relapsed plasma cell myeloma: an NCI (Canada) pilot study. Br J Haematol 1992; 82:555-9.

Bruno B, Rotta M, et al.(2007) A comparison of allografting with autografting for newly diagnosed myeloma. NEJM, 2007; 356:110-20.

Bruno B, Rotta M, et al.(2009) Nonmyeloablative allografting for newly diagnosed multiple myeloma: the experience of the Gurppi Italiano Trapianti di Midollo. Blood, 2009; 113"3375-82.

Cavallo F, Ambrosini MT, et al.(2007) The treatment of the elderly multiple myeloma patients. Leukemia & Lymphoma, 2007; 48(3):469-80.

Cavo M, Gay F, Beksac M, et al.(2020) Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a multicentre, randomised, open-label, phase 3 study. Lancet Haematol. Jun 2020; 7(6): e456-e468. PMID 32359506

Cavo M, Tosi P, et al.(2007) Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. J Clin Oncol, 2007; 25(17):2434-41.

Child JA, Morgan GJ, Davies FE, et al.(2003) High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. NEJM 2003; 348: 1875-1883.

Clinicaltrials.gov.(2020) Continued, Long-Term Follow-Up and Lenalidomide Maintenance Therapy for Patients on BMT CTN 0702 Protocol (BMT CTN 07LT). Updated May 11, 2020. Accessed November 18, 2021. https://clinicaltrials.gov/ct2/show/NCT02322320

Cook G, Ashcroft AJ, Cairns DA, et al.(2016) The effect of salvage autologous stem-cell transplantation on overall survival in patients with relapsed multiple myeloma (final results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a randomised, open-label, phase 3 trial. Lancet Haematol. Jul 2016;3(7):e340-351. PMID 27374467

Cook G, Williams C, Brown JM, et al.(2014) High-dose chemotherapy plus autologous stem-cell transplantation as consolidation therapy in patients with relapsed multiple myeloma after previous autologous stem-cell transplantation (NCRI Myeloma X Relapse [Intensive trial]): a randomised, open-label, phase 3 trial. Lancet Oncol. Jul 2014;15(8):874-885. PMID 24948586

Crawley C, Iacobelli S, et al.(2007) Reduced-intensity conditioning for myeloma: lower nonrelapse mortality but higher relapse rates compared with myeloablative conditioning. Blood, 2007; 109:3588-94.

Cunningham D, Paz-Ares L, Milan S, et al.(1994) High-dose autologous bone marrow transplantation as consolidation in previously untreated myeloma. J Clin Oncol 1994; 12:759-63.

Delain M, Linassier C, Petitdidier C, et al.(1994) VAD-PECC regimen in the treatment of advanced-stage multiple myeloma. J Clin Oncol 1994; 12:2706-13.

Dhakal B, Shah N, Kansagra A, et al.(2022) ASTCT Clinical Practice Recommendations for Transplantation and Cellular Therapies in Multiple Myeloma. Transplant Cell Ther. Jun 2022; 28(6): 284-293. PMID 35306217

Dimopoulos MA, Hester J, Huh Y, et al.(1994) Intensive chemotherapy with blood progenitor transplantation for primary resistant multiple myeloma. Br J Hematol 1994; 87:730-4.

Dimopoulos MA, Weber DM, Hester J, et al.(1994) Intensive sequential therapy for VAD-resistant multiple myeloma. Leuk Lymphoma 1994; 13:479-84.

Dispenzieri A, Lacy MQ, Kyle RA, et al.(2001) Eligibility for hematopoietic stem-cell transplantation for primary systemic amyloidosis is a favorable prognostic factor for survival. J Clin Oncol 2001;19:3350-56.

Dunavin NC, Wei L, Elder P, et al.(2013) Early versus delayed autologous stem cell transplant in patients receiving novel therapies for multiple myeloma. Leuk Lymphoma. Aug 2013;54(8):1658-1664. PMID 23194056

Durie BG, Harousseau JL, Miguel JS, et al.(2006) International uniform response criteria for multiple myeloma. Leukemia. Sep 2006; 20(9): 1467-73. PMID 16855634

Farina L, Bruno B, et al.(2009) The hematopoiectic cell transplantation comorbidity index (HCT-CI) predicts clinical outcomes in lymphoma and myeloma patients after reduced-intensity or non-myeloablative allogeneic stem cell transplantation. Leukemia, 2009; 23(6):1131-8.

Fermand JP, Chevret S, Levy Y, et al.(1992) The role of autologous blood stem cells in support of high-dose therapy for multiple myeloma. Hematol Oncol Clin N Am 1992; 6:451-62.

Fermand JP, Chevret SS, Ravaud P, et al.(1993) High dose chemoradiotherapy and autologous blood stem cell transplantation in multiple myeloma: results of a phase II trial involving 63 patients. Blood 1993; 82:2005-9.

Fermand JP, Ravaud P.(1997) High dose therapy (HDT) and autologous peripheral blood stem cell (PBSC) transplantation performed either as first-line therapy or as a rescue treatment: similar effect on overall survival in myeloma patients. VI International Workshop on Multiple Myeloma 1997; Boston; Harvard Med School and Dana-Farber Cancer Institute.

Gahrton G, Iacobelli S, Bjorkstrand B, et al.(2013) Autologous/reduced-intensity allogeneic stem cell transplantation vs autologous transplantation in multiple myeloma: long-term results of the EBMTNMAM2000 study. Blood. Jun 20 2013;121(25):5055-5063. PMID 23482933

Ganjoo RK, Williams A, Malpas JS.(1995) Vincristine and oral etoposide in refractory multiple myeloma. Cancer Chemother Pharmacol 1995; 35:343-4.

Gay F, Oliva S, Petrucci MT, et al.(2015) Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol. Dec 2015;16(16):1617-1629. PMID 26596670

Gertz MA, Garton JP, Greipp PR, et al.(1995) A phase II study of high dose methylprednisolone in refractory or relapsed multiple myeloma. Leukemia 1995; 9:2115-8.

Gertz MA, Kalish LA, Kyle RA, et al.(1995) Phase III study comparing vincristine, doxorubicin (Adriamycin), and dexamethasone (VAD) chemotherapy with VAD plus recombinant interferon alfa-2 in refractory or relapsed multiple myeloma. Am J Clin Oncol 1995; 18:475-80.

Gertz MA, Lacy MQ, Dispenzieri A.(2000) Myeloablative chemotherapy with stem cell rescue for the treatment of primary systemic amyloidosis: a status report. BMT 2000; 25: 465-70.

Gertz MA, Pineda AA, Chen MG, et al.(1995) Refractory and relapsing multiple myeloma treated by blood stem cell transplantation. Am J Med Sci 1995; 309:152-61.

Gianni AM, Tarella C, Bregni M, et al.(1994) High-dose sequential chemoradiotherapy, a widely applicable regimen, confers survival benefit to patients with high-risk multiple myeloma. J Clin Oncol 1994; 12:503-9.

Giralt S, Costa L, Schriber J, et al.(2014) Optimizing autologous stem cell mobilization strategies to improve patient outcomes: consensus guidelines and recommendations. Biol Blood Marrow Transplant. Mar 2014;20(3):295- 308. PMID 24141007

Giralt S, Garderet L, Durie B, et al.(2015) American Society of Blood and Marrow Transplantation, European Society of Blood and Marrow Transplantation, Blood and Marrow Transplant Clinical Trials Network, and International Myeloma Working Group consensus conference on salvage hematopoietic cell transplantation in patients with relapsed multiple myeloma. Biol Blood Marrow Transplant. Dec 2015;21(12):2039-2051. PMID 26428082

Giralt S, Koehne G.(2013) Allogeneic hematopoietic stem cell transplantation for multiple myeloma: what place, if any? Curr Hematol Malig Rep. Dec 2013;8(4):284-290. PMID 24146203

Giralt S, Weber D, Colome M, et al.(1997) Phase I trial of cyclosporine-induced autologous graft-versus-host disease in patients with multiple myeloma undergoing high-dose chemotherapy with autologous stem-cell rescue. J Clin Oncol 1997; 15:667-73.

Goldschmidt H, Baertsch MA, Schlenzka J, et al.(2020) Salvage autologous transplant and lenalidomide maintenance vs. lenalidomide/dexamethasone for relapsed multiple myeloma: the randomized GMMG phase III trial ReLApsE. Leukemia. Jul 21 2020. PMID 32694619

Goldschmidt H, Hegenbart U, Wallmeier M, et al.(1997) High-dose therapy with peripheral blood progenitor cell transplantation in multiple myeloma. Ann Oncol 1997; 8:243-246.

Gregory W, Richards MA, Malpas JS.(1992) Combination chemotherapy versus melphalan and prednisolone in the treatment of multiple myeloma: an overview of published trials. J Clin Oncol 1992; 10:334-42.

Greipp PR, Lust JA, O’Fallon WM, et al.(1993) Plasma cell labeling index and beta 2-microglobulin predict survival independent of thymidine kinase and C-reactive protein in multiple myeloma. Blood 1993; 81:3382-7.

Harousseau JL, Attal M, Divine M, et al.(1995) Autologous stem cell transplantation after first remission induction treatment in multiple myeloma: a report of the French Registry on Autologous Transplantation in Multiple Myeloma. Blood 1995; 85:3077-85.

Harousseau JL, Milpied N, Laporte JP, et al.(1992) Double-intensive therapy in high-risk multiple myeloma. Blood 1992; 79:2827-33.

Harousseau JL, Moreau P.(2007) Role of bone marrow transplantation in the disease pathway of myeloma. J Natl Compr Canc Netw, 2007; 5(2):163-9.

Harousseau JL, Moreau P.(2009) Autologous hematopoietic stem-cell transplantation for multiple myeloma. NEJM, 2009; 360:2645-54.

Hawkins T, Horvath N, Rawling C, et al.(1996) An incremental response to high dose therapy in multiple myeloma. BMT 1996; 17:929-35.

Henon P, Donatini B, Eisenmann JC, et al.(1995) Comparative survival, quality of life and cost-effectiveness of intensive therapy with autologous blood cell transplantation or conventional chemotherapy in multiple myeloma. BMT 1995; 16:19-25.

Ikeda T, Mori K, Kawamura K, et al.(2019) Comparison between autologous and allogeneic stem cell transplantation as salvage therapy for multiple myeloma relapsing/progressing after autologous stem cell transplantation. Hematol Oncol. Dec 2019; 37(5): 586-594. PMID 31674032

Jagannath S, Vesole DH, Glenn L, et al.(1992) Low-risk intensive therapy for multiple myeloma with combined autologous bone marrow and blood stem-cell support. Blood 1992; 80:1666-72.

Jagannath S, Vesole DH, Tricot G, et al.(1994) Hemopoietic stem cell transplants for multiple myeloma. Oncology 1994; 8:89-103.

Jagannath S, Vesole DH, Zhang M, et al.(1997) Feasibility and cost-effectiveness of outpatient autotransplants in multiple myeloma. BMT 1997; 20:445-450.

Jang IY, Yoon DH, Kim S, et al.(2014) Advanced POEMS syndrome treated with high-dose melphalan followed by autologous blood stem cell transplantation: a single-center experience. Blood Res. Mar 2014;49(1):42-48. PMID 24724066

Johnson RJ, Owen RG, Smith GH, et al.(1996) Peripheral blood stem cell transplantation in myeloma using CD34 selected cells. BMT 1996; 17:723-7.

Kanate AS, Majhail NS, Savani BN, et al.(2020) Indications for Hematopoietic Cell Transplantation and Immune Effector Cell Therapy: Guidelines from the American Society for Transplantation and Cellular Therapy. Biol Blood Marrow Transplant. Jul 2020; 26(7): 1247-1256. PMID 32165328

Koreth J, Cutler CS, et al.(2007) High-dose therapy with single autologous transplantation for chemotherapy for newly diagnosed multiple myeloma: a systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant, 2007; 13(183-96.

Kraut EH, Crowley JJ, Wade JL, et al.(1998) Evaluation of topotecan in resistant and relapsing multiple myeloma: a Southwest Oncology Group study. J Clin Oncol 1998; 16:589-92.

Krishnan A, Stadtmauer E.(2010) Treatment -related death toll casts doubt on "auto-allo" transplants in myeloma. http://ash.confex.com/ash/2010/webprogram/Paper30659.html

Kroger N.(2007) Mini-Midi-Maxi? How to harness the graft-versus-myeloma effect and target molecular remission after allogeneic stem cell transplantation. Leukemia, 2007; 21(9):1851-8.

Kumar A, Kharfan-Dabaja MA, et al.(2009) Tandem versus single autologous hematopoietic cell transplantation for the treatment of multiple myeloma: a systematic review and meta-analysis. J Natl Cancer Inst, 2009; 101:100-6.

Kyle RA, Rajkumar SV.(2008) Multiple myeloma. Blood. Mar 15 2008; 111(6): 2962-72. PMID 18332230

Kyle RA, Rajkumar SV.(2009) Treatment of multiple myeloma: a comprehensive review. Clin Lymphoma Myeloma, 2009; 9(4):278-88.

Kyle RA.(1999) High dose therapy in multiple myeloma and primary amyloidosis: An overview. Semin Oncol 1999; 26:74-83.

Lenhard RE, Daniels MJ, Oken MM, et al.(1994) An aggressive high dose cyclophosphamide and prednisone regimen for advanced multiple myeloma. Leuk Lymphoma 1994; 13:485-9.

Lenhard RE, Kalish LA, Oken MM, et al.(1994) Timed sequential high-dose cyclophosphamide and vincristine in the treatment of multiple myeloma. Cancer 1994; 73:2113-8.

Levenga H, Levenga H, Levison-Keating S, et al.(2007) Multiple myeloma patients receiving pre-emptive donor lymphocytic infusion after patient T-cell-depleted allogeneic stem cell transplantation show a long progression-free survival. Bone Marrow Transplant, 2007; 40(4):355-9.

Lokhorst H, Einsele H, Vesole D, et al.(2010) International Myeloma Working Group consensus statement regarding the current status of allogeneic stem-cell transplantation for multiple myeloma. J Clin Oncol. Oct 10 2010;28(29):4521-4530. PMID 20697091

Long G, Chao N, Hu W, et al.(1996) High dose etoposide-based myeloablative therapy followed by autologous blood progenitor cell rescue in the treatment of multiple myeloma. Cancer 1996; 78:2502-9.

Mahendra P, Hood IM, Johnson D, et al.(1995) Autografting for multiple myeloma: a 5-year experience at a single institution. BMT 1995; 16:759-63.

Majolino I, Davoli M, et al.(2007) Reduced intensity conditioning with thiotepa, fludarabine, and melphalan is effective in advanced mutliple myeloma. Leuk Lymphoma, 2007; 48(4):759-66.

Mansi JL, Cunningham D, Viner C, et al.(1993) Repeat administration of high dose melphalan in relapsed myeloma. Br J Cancer 1993; 68:983-7.

Marit G, Faberes JL, Pico JM, et al.(1996) Autologous peripheral-blood progenitor-cell support following high dose chemotherapy or chemoradiotherapy in patients with high-risk multiple myeloma. J Clin Oncol 1996; 14:1306-1313.

McCarthy PL, Holstein SA.(2016) Role of stem cell transplant and maintenance therapy in plasma cell disorders. Hematology Am Soc Hematol Educ Program. Dec 2 2016;2016(1):504-511. PMID 27913522

Mehta J, Tricot G, Jagannath S, et al.(1997) High-dose chemotherapy with carboplatin, cyclophosphamide and etoposide and autologous transplantation for multiple myeloma relapsing after a previous transplant. BMT 1997; 20:113-6.

Mehta J.(2009) Re: Tandem vs signle autologous hematopoietic cell trasplanation for the treatment of multiple myeloma: a systematic review and meta-analysis. J Natl Canc Inst, 2009; 101(20):1430-1.

Mian H, Mian OS, Rochwerg B, et al.(2020) Autologous stem cell transplant in older patients (age65) with newly diagnosed multiple myeloma: A systematic review and meta-analysis. J Geriatr Oncol. Jan 2020; 11(1): 93-99. PMID 31153809

Michaelis LC, Saad A, Zhong X, et al.(2013) Salvage second hematopoietic cell transplantation in myeloma. Biol Blood Marrow Transplant. May 2013;19(5):760-766. PMID 23298856

Mikhael J, Ismaila N, Cheung MC, et al.(2019) Treatment of Multiple Myeloma: ASCO and CCO Joint Clinical Practice Guideline. J Clin Oncol. May 10 2019; 37(14): 1228-1263. PMID 30932732

National Comprehensive Cancer Network (NCCN).(2021) NCCN Clinical Practice Guidelines in Oncology: Multiple Myeloma. Version 3.2022. Accessed November 18, 2021. https://www.nccn.org/professionals/physician_gls/pdf/myeloma.pdf

Palumbo A, Pileri A, Tricolo S, et al.(1997) Multicyclic, dose-intensive chemotherapy supported by hemopoietic progenitors in refractory myeloma patients. BMT 1997; 19:23-29.

Palumbo A, Rajkumar SV.(2009) Treatment of newly diagnosed myeloma. Leukemia. Mar 2009; 23(3): 449-56. PMID 19005483

Phillips JK, Sherlaw-Johnson C, Pearce R, et al.(1995) A randomized study of MOD versus VAD in the treatment of relapsed and resistant multiple myeloma. Leuk Lymphoma 1995; 17:465-72.

Powles R, Raje N, Milan S, et al.(1997) Outcome assessment of a population-based group of 195 unselected myeloma patients under 70 years of age offered intensive treatment. BMT 1997; 20:435-443.

Qiao SK, Guo XN, Ren JH, et al.(2015) Efficacy and Safety of Lenalidomide in the Treatment of Multiple Myeloma: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Chin Med J (Engl). 5th May 2015;128(9):1215-1222. PMID 25947406

Raje N, Powles R, Horton C, et al.(1997) Comparison of marrow versus blood-derived stem cells for autografting in previously untreated multiple myeloma. Br J Cancer 1997; 75:1684-1689.

Rajkumar SV, Kumar S.(2020) Multiple myeloma current treatment algorithms. Blood Cancer J. Sep 28 2020; 10(9): 94. PMID 32989217

Rajkumar SV.(2020) Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am J Hematol. May 2020; 95(5): 548-567. PMID 32212178

Ramos CA, Saliba RM, et al.(2009) Impact of hepatitis C virus serpositivity on survival after allogeneic hematopoietic stem cell transplantation for hematologic malignancies. Haematoligica, 2009; 24(2):249-57.

Reece D HJ, Gertz MA.(2009) Myeloma Management 2009: Nontransplant therapy of myeloma, high-dose therapy for myeloma, and a personalised care plan for treatment of myeloma. 2009 American Society of Clinical Oncology Annual Meeting Educational Handbook. 2009:502-509.

Reece DE.(2009) Recent trends in the management of newly diagnosed multiple myeloma. Curr Opin Hematol. Jul 2009; 16(4): 306-12. PMID 19491669

Richardson PG, Jacobus SJ, Weller EA, et al.(2022) Triplet Therapy, Transplantation, and Maintenance until Progression in Myeloma. N Engl J Med. Jul 14 2022; 387(2): 132-147. PMID 35660812

Rosinol L, Perez-Simon JA, et al.(2008) A prospective (PETHEMA study of tandem autologous transplantation versus autograft followed by reduced-intensity conditioning allogeneic transplantation in newly diagnosed multiple myeloma. Blood, 2008; 112(9):3591-3.

Rotta M, Storer BE, et al.(2009) Long-term outocme of patients with multiple myeloma after autologous hematopoietic cell transplantation and nonmyeloablative allografting. Blood, 2009; 113(14):3383-91.

Salmon SE, Cassady JR.(1993) Plasma cell neoplasms. Cancer:Principles and Practice ncology. 4th ed. JB Lippincott Co.; 1984-2025. DeVita VT et al, eds; 1993.

Schiller G, Vescio R, Freytes C, et al.(1995) Transplantation of CD34+ peripheral blood progenitor cells after high-dose chemotherapy for patients with advanced multiple myeloma. Blood 1995; 86:390-7.

Schmidt-Hieber M, Blau IW, et al.(2007) Reduced-toxicity conditioning with fludarabine and treosulfan prio to allogeniec stem cell transplantation in multiple myeloma. Bone Marrow Transplant, 2007; 39(7):389-96.

Shah N, Callander N, Ganguly S, et al.(2015) Hematopoietic Stem Cell Transplantation for Multiple Myeloma: Guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. Mar 11 2015. PMID 25769794

Stadtmauer EA, Pasquini MC, Blackwell B, et al.(2019) Autologous Transplantation, Consolidation, and Maintenance Therapy in Multiple Myeloma: Results of the BMT CTN 0702 Trial. J Clin Oncol. Mar 01 2019; 37(7): 589-597. PMID 30653422

Tricot G, Jagannath S, Vesole D, et al.(1995) Peripheral blood stem cell transplants for multiple myeloma: identification of favorable variables for rapid engraftment in 225 patients. Blood 1995; 85:588-96.

Tricot G, Jagannath S, Vesole D, et al.(1995) Relapse of multiple myeloma after autologous transplantation: survival after salvage therapy. BMT 1995; 16:7-11.

Tricot G, Kern SE, Barlogie B.(2009) Re: Tandem vs signle autologous hematopoietic cell trasplanation for the treatment of multiple myeloma: a systematic review and meta-analysis. J Natl Canc Inst, 2009; 101(13):964-6.

Tricot G, Sawyer JR, Jagannath S, et al.(1997) Unique role of cytogenetics in the prognosis of patients with myeloma receiving high-dose therapy and autotransplants. J Clin Oncol 1997; 15:2659-2666.

Varterasian M, Janakiraman N, Karanes C, et al.(1997) Transplantation in patients with multiple myeloma: a multicenter comparative analysis of peripheral blood stem cell and allogeneic transplant. Am J Clin Oncol 1997; 20:462-466.

Vesole DH, Barlogie B, Jagannath S, et al.(1994) High-dose therapy for refractory multiple myeloma: improved prognosis with better supportive care and double transplants. Blood 1994; 84:950-6.

Vesole DH, Zhang L, et al.(2009) A phase II trial of autologous stem cell transplantation followed by mini-allogeneic stem cell transplantation for the treatment of multiple myeloma: an analysis of Eastern Cooperative Oncology Group ECOG E4A98 and E1A97. Biol Blood Marrow Transplant, 2009; 15:83-91.

Villalba A, Gonzalez-Rodriguez AP, Arzuaga-Mendez J, et al.(2022) Single versus tandem autologous stem-cell transplantation in patients with newly diagnosed multiple myeloma and high-risk cytogenetics. A retrospective, open-label study of the PETHEMA/Spanish Myeloma Group (GEM). Leuk Lymphoma. Sep 20 2022: 1-10. PMID 36124538

Weber D, Dimopoulos M, Sinicrope F, et al.(1995) VAD-cyclosporine therapy for VAD-resistant multiple myeloma. Leuk Lymphoma 1995; 19:159-63.

Ziogas DC, Terpos E, Dimopoulos MA.(2017) When to recommend a second autograft in patients with relapsed myeloma? Leuk Lymphoma. Leuk Lymphoma. Apr 2017;58(4):781-787. PMID 27894207


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.