• Non ci sono risultati.

Nonmyeloablative Stem Cell Transplantation in the Treatment of Hematologic Malignancies 25

N/A
N/A
Protected

Academic year: 2022

Condividi "Nonmyeloablative Stem Cell Transplantation in the Treatment of Hematologic Malignancies 25"

Copied!
10
0
0

Testo completo

(1)

Allogeneic hematopoietic stem cell transplanta- tion (SCT) is an effective, potentially curative treatment of advanced or high-risk hematologic malignancies.1 High-dose chemoradiotherapy with allogeneic SCT is associated with signifi- cant morbidity and mortality because of the toxicity of the preparative regimen, graft-versus- host disease (GVHD), and the immune-deficiency state that accompanies the procedure. These risks are significantly increased with advanced age, concurrent medical problems, or extensive prior therapy, limiting standard SCT to younger patients in good medical condition. Hematologic malignancies are more common and have a worse prognosis in the elderly. Additionally, dis- ease and prior therapy may result in comorbidi- ties precluding further intensive therapy. Thus, many patients with hematologic malignancies who could benefit from SCT were often deferred from a potentially curative approach. Extensive research has been directed toward the develop- ment of safer and less toxic approaches to allogeneic SCT. The introduction of nonmye- loablative and reduced-intensity conditioning regimens is a major step toward extension of allogeneic SCT to a much wider patient popula- tion by reducing transplant-related complica- tions.2 Much experience has been gained with the clinical use of this novel approach over the last decade. In this chapter, we discuss the rationale for nonmyeloablative stem cell trans- plantation (NST), and the use of immune thera- peutic interventions with NST as the curative approach. We discuss how NST reduces some,

but not all transplant-related complications, and our personal approach in selecting patients for NST.

Rationale for NST

SCT was initially developed as a means to deliver high-dose chemotherapy and radiation for elimination of the underlying disorder.

Escalation of treatment doses results in better tumor kill but leads to irreversible myelosup- pression. SCT was viewed as a supportive-care modality to restore hematopoiesis after treat- ment. However, it has subsequently become apparent that high-dose chemoradiotherapy does not eradicate the disease in many patients and that much of the therapeutic benefit of SCT relates to an associated, immune-mediated, graft-versus-leukemia (GVL) or graft-versus- malignancy (GVM) effect. Extensive experimen- tal and clinical data support the presence of this GVL effect.2Higher relapse rates were observed after syngeneic and T cell-depleted transplants whereas patients having acute or chronic GVHD have a reduced relapse risk suggesting the importance of T cell-mediated immunity in eliminating the malignancy and the association of this GVL effect with GVHD. Perhaps the most direct evidence for GVL/GVM was the ability to restore remissions in patients relapsing after SCT by infusion of donor lymphocytes with no additional chemotherapy. The discovery of the

25

Nonmyeloablative Stem Cell Transplantation in the Treatment of Hematologic Malignancies

Avichai Shimoni and Arnon Nagler

351

(2)

curative potential of the immune-mediated GVL/GVM effect has led to a novel therapeutic approach. Low-dose, relatively nontoxic and tolerable conditioning regimens have been designed, not to eradicate the malignancy, but rather to provide sufficient immunosuppression to achieve donor cell engraftment and to allow induction of GVL as the primary treatment.2–4

NST does not eliminate all host hematopoiesis and often leads to a state of mixed chimerism (MC). MC describes persistence of donor cells with either normal host hematopoietic cells and/or cells of the underlying malignancy (Figure 25.1).

Stable long-lived MC has been reported in ani- mal models and in patients having NST for non- malignant disorders. However, in patients with malignancies, MC is most often transient and conversion to complete chimerism (CC), autolo- gous reconstitution, or relapse occurs either spontaneously or after immune manipulations within the first few months after NST.5The ini- tial nonmyeloablative treatment is expected to produce only transient suppression of the underlying malignancy, but it allows time for the immune GVM effect to develop. This effect may result in gradual elimination of the malig- nancy and spontaneous delayed achieve- ment of complete remission (CR), over a few months, especially in indolent malignancies.

However, patients with MC or with detectable residual malignancy after NST may require additional immune-therapeutic approaches.

Immunosuppressive therapy given post-SCT for prevention of GVHD can also suppress the GVL effect.6Early withdrawal of immunosup- pressive therapy allows the occurrence of potent graft-versus-hematopoietic tissue effect that can potentially eliminate both residual disease and

host hematopoiesis producing CC and CR (Figure 25.1). If this does not occur, donor lym- phocyte infusions (DLIs) may harness this effect and switch the balance toward CC/CR. The GVL and graft-versus-hematopoietic tissue effects are highly associated with GVHD although may also occur in its absence. The initial NST and donor cell engraftment thus serve as a platform for additional allogeneic cellular therapy.

NST Regimens

NST regimens comprise a spectrum of regimens with different immunosuppressive and myelo- suppressive properties. The kinetics of engraft- ment, chimerism, and eradication of residual disease differ accordingly.5 Conditioning regi- mens have been referred to as nonmyeloabla- tive if they do not completely eradicate host hematopoiesis and immunity.2 A few of these regimens have been given as chemotherapeutic regimens with no stem cell support and allow relatively prompt hematologic recovery.

Autologous reconstitution of hematopoiesis is expected if the allograft is rejected. These non- myeloablative regimens have potent immuno- suppressive effects. They are only mildly myelosuppressive and often result in induction of MC. The Seattle regimen consisting of low- dose total body irradiation (TBI, 200 cGy) with (or initially without) fludarabine and intensive pre- and posttransplant immunosuppression is the prototype of these regimens.7Other exam- ples are the combinations of fludarabine and cyclophosphamide (FC) and the Flag/Ida regi- men developed initially at the MD Anderson for

m m

Nonmyeloablative conditioning

IST withdrawal

±DLI

recipient donor mixed chimerism

+MRD

complete chimerism

Figure 25.1. NST program: The initial NST regimen induces MC with persistence of both donor and recipient hematopoietic cells. The underlying malignancy (m) is suppressed but not completely eliminated. In the second phase, immune-therapeutic interventions, e.g., withdrawal of immuno- suppressive therapy (IST) supplemented if necessary by DLI, induce graft-versus-hematopoietic tissue and graft-versus-tumor effects eliminating recipient hematopoiesis and the underlying malignancy and converting to CC.

(3)

non-SCT treatment of lymphoid and myeloid malignancies, respectively, and later explored as nonmyeloablative conditioning regimens for SCT.4,8These are very tolerable regimens, allow- ing in some cases ambulatory treatment, and treatment of elderly patients.

More intensive regimens have also been developed. These regimens have been referred to as reduced-intensity conditioning regimens.2 They have not been given without stem cell sup- port, and autologous recovery after treatment may be slow if at all. These regimens usually combine immunosuppressive agents such as flu- darabine with or without serotherapy [with anti- thymocyte globulin (ATG) or alemtuzumab]

and agents with moderate myelosuppressive effects (such as busulfan or melphalan).3,9,10The Hadassah group3and the MD Anderson group4 pioneered the use of purine analogs in NST reg- imens and they emerged as the cornerstone of these regimens. These are well-tolerated agents, with potent immunosuppressive effects, in addi- tion to antitumor activity against a range of hematologic malignancies. They have synergistic effects with alkylating agents and inhibit DNA repair systems responsible for repair of cellular damage induced by these agents. Although these regimens are more intensive than the nonmye- loablative regimens, dose intensity is still reduced compared with standard ablative regi- mens allowing reduction of toxicity. Reduced- intensity regimens, in similarity to standard myeloablative regimens, rapidly induce CC and antitumor responses, but are more toxic, and associated with a higher risk for GVHD.5

A third approach is using a double-step strategy. Initially, high-dose chemotherapy sup- ported by autologous stem cell transplanta- tion is used for cytoreduction and also as an immunosuppressive platform for the second stage of allogeneic SCT with nonmyeloablative or reduced-intensity regimens usually adminis- tered 2–3 months later. The separation of high- dose chemotherapy and allogeneic effects results in reduced toxicity and better tolerability than when allogeneic transplantation immedi- ately follows high-dose chemotherapy.11,12

A novel approach is to combine nonmyeloab- lative or reduced-intensity regimens with tar- geted therapy. Imatinib is being explored as adjuvant to reduced-intensity conditioning both pre-SCT, allowing reduction of conditioning intensity, and post-SCT, to eliminate MRD.13 Rituximab has been used in conjunction with

reduced-dose chemotherapy in lymphoid malig- nancies, and by us after SCT to target MRD.14 More recently, radiolabeled immune conjugates are used with SCT. Antibodies such as radiola- beled anti-CD20 monoclonal antibodies may be used with SCT to target lymphoma cells allow- ing the use of less intensive conditioning.

Radiolabeled antibodies such as Bismuth 213 anti-CD45 antibodies can be used with no addi- tional chemotherapy to ablate the marrow and immune system and not specifically tumor cells as reported in preliminary canine models.15

NST and Regimen-Related Complications

NST regimens were originally designed to enable treatment of older and medically infirm patients not eligible for standard ablative condi- tioning and to allow the application of SCT to a much wider patient population. This goal has largely been achieved. Standard ablative regi- mens are usually limited to patients up to age 55 years. Most NST studies have no upper age limit.

Age per se was not found to be an adverse factor for prediction of outcome16,17after both related and unrelated donor SCT and is no longer a con- traindication for SCT. Standard SCT in certain high-risk settings such as in heavily pretreated patients, patients failing a prior autologous SCT, and in patients with certain diagnoses such as multiple myeloma, Hodgkin’s and non-Hodgkin’s lymphoma, was associated with unacceptably high treatment-related mortality (TRM) rates, as high as 50%. TRM in the range of 10%–20% can now be observed in these settings using NST regimens. In particular, NST is becoming a com- mon indication for treatment of patients failing a prior autologous SCT,18,19 and was able to reduce TRM after unrelated donor SCT.20,21

Reduction of TRM is largely attributed to reduction in organ toxicity. The Seattle group has shown marked reduction in cardiovascular, gastrointestinal, hepatic, infectious, metabolic, neurologic, and pulmonary toxicity when com- paring their low-dose TBI-based nonmyeloabla- tive regimen to ablative regimens.22Nonrelapse mortality within the first 100 days was 9% and 21%, respectively. The major therapy-related organ dysfunction syndromes are reduced in incidence. In particular, idiopathic pneumonia

(4)

syndrome is less frequent after NST, 2.2% versus 8.4% in one study, despite treatment of older patients.23Hepatic toxicity may still be substan- tial, especially after some reduced-intensity reg- imens.3,24 However, not all syndromes are reduced. We have shown that thrombotic microangiopathy is a frequent devastating com- plication after NST, more common in second SCTs and in association with acute GVHD.25 Diffuse alveolar hemorrhage is also relatively common in this setting. We have hypothesized based on experimental data that fludarabine- related endothelial and pulmonary epithelial toxicity may be associated with this unexpected observation. It was also shown that other hemolytic complications, associated with ABO donor–recipient incompatibility, might be more common after NST.26Although direct toxicities of high-dose chemotherapy are reduced with NST, toxicities involving immune mechanisms may not be. Organ toxicities are largely associ- ated with patient comorbidity score before SCT.27Further research is required to define the relative organ toxicities in different regimens.

NST is less myelosuppressive than standard conditioning. This results in a shorter duration of neutropenia and less transfusion require- ments.28 Some of the nonmyeloablative regi- mens result in only minimal neutropenia and can be safely administered in the outpatient set- ting.7Reduced-intensity regimens usually result in more profound cytopenias more similar to ablative conditioning. The reduced duration of neutropenia and the limitation of mucosal injury result in reduced risk for severe infections in the immediate post-SCT period.29,30However, the risk for invasive fungal infections is not reduced.31,32These infections are usually associ- ated with GVHD and corticosteroid therapy, and represent one of the major causes of TRM after NST. In the Seattle study, invasive fungal infection occurred in 19% of NST recipients, they occurred relatively late in the course, at a median of 107 days, and were the primary cause for 39% of nonrelapse-associated deaths.31

NST and GVHD

GVHD is one of the major causes of post-SCT morbidity and mortality. When the nonmye- loablative and reduced-intensity regimens were introduced, it was hoped that GVHD incidence

would reduce. Acute GVHD results at least par- tially from tissue injury and cytokine release secondary to the toxicity of the preparative regimen, amplified by donor immune cells.33 The use of less toxic conditioning should theo- retically limit tissue injury and cytokine release and reduce the incidence and severity of GVHD.

Also, MC that is more common after NST allows bilateral transplantation tolerance with graft acceptance and some protection from GVHD.34 However, host antigen-presenting cells that have a major role in initiation of the GVHD reaction may persist after NST and contribute to GVHD.35 The duration of immunosuppres- sive therapy is usually shorter after NST, and immune manipulations are often incorpo- rated into NST programs increasing the likeli- hood of GVHD although delayed immune manipulations, once the toxicity of condition- ing and cytokine release are already resolved, are less likely to produce severe GVHD.36 The net effect of these differences between NST and ablative SCT on GVHD is still not well estab- lished and is controversial. The Seattle group reported that the incidence of acute GVHD grade II–IV after NST was significantly lower than after ablative therapy, reaching 64% and 85%, respectively. However, the incidence of chronic GVHD was approximately 70% in both cohorts. Moreover, the initiation of steroid ther- apy was delayed from an average of 1 month to 3 months after SCT, corresponding to a “new”

syndrome described as late-onset acute GVHD.37This study suggests that GVHD is not reduced in incidence with NST, but is only delayed. In another study, Couriel et al.38 reported an incidence of grade II–IV acute GVHD of 36% after myeloablative regimens (including the reduced-intensity combination of fludarabine and melphalan), but only 12% after truly nonmyeloablative regimens. They also noted reduced incidence of chronic GVHD after NST.37 Further prospective studies are needed to deter- mine the relative incidence of GVHD. However, Because it is still a major cause of morbidity and mortality after NST, several approaches have been explored to decrease the risk.

Initially, NST regimens called for only a short course of immune suppression and early admin- istration of DLI for disease eradication and con- version to CC. However, these interventions are thought to markedly increase the risk of GVHD. More recently, more careful approaches were introduced. For example, the Seattle group

(5)

extended the duration of immune suppression, especially after unrelated donor transplantation, up to 6 months. With better understanding of chimerism and MRD kinetics, the indications for DLI have been restricted, trying to reserve it only for patients destined to relapse or reject their graft and reducing the risk of GVHD in all other SCT recipients (see below for further dis- cussion).

Another approach is the use of in vivo T cell depletion. Alemtuzumab (Campath 1-H) has emerged as an effective agent in prevention of GVHD. Alemtuzumab administered during pre- SCT conditioning depletes host T cells thus reducing the risk of graft rejection reported with in vitro T cell depletion techniques. Alemtuzumab persists after SCT and also depletes, at least par- tially, T cells of the donor, as well as host antigen- presenting cells and thus has been shown to be very effective in prevention of GVHD after reduced-intensity conditioning from both related and unrelated donors. However, patients given alemtuzumab have a higher risk of opportunistic infections, in particular with cytomegalovirus.

Moreover, alemtuzumab recipients have a higher risk of MC and residual disease, require more DLIs, such that after DLI, the ultimate net risk of GVHD is not reduced and there is no improve- ment in survival or TRM.10ATG given pre-SCT has the same effects although may be less effec- tive in prevention of GVHD. Studies are being conducted to determine the dose of alem- tuzumab or ATG that may result in net effects that would improve survival.

Immune-Therapeutic Intervention after NST

Relapse of the underlying malignancy remains the major cause of treatment failure after abla- tive SCT and even more so after NST. Most of the data on the safety and efficacy of DLI comes from myeloablative SCT. DLI has been admin- istered after NST in a variety of indications, mostly for conversion of MC to CC and for the treatment of relapse or residual disease.39DLI is associated with significant morbidity and mor- tality, mostly because of complications related to GVHD and marrow aplasia. Marks et al.39 reported in a large series of DLI after NST that the rate of severe GVHD was 15%, and TRM was 9%; marrow aplasia was rare, suggesting that

DLI after NST was safer than what is reported after standard myeloablative SCT. The Seattle group reported similar results.40This may repre- sent advances in DLI administration, such as administration in incremental dosing, and at MRD where DLI may also be more effective, and may be given at a lower starting dose. There is also experimental data suggesting that DLI may even be more potent in mixed chimeras because of persistence of host antigen-presenting cells.40 DLI administered late after SCT has a lower risk of complications36; however, the window of opportunities for administration of DLI for pre- vention of relapse may be short and missed while waiting for a safer time point. Even in pro- grams planning early DLI, on days 60–100 post- SCT, DLI is only administered after cyclosporine withdrawal and many patients are ineligible for prophylactic DLI because of GVHD or rapid progression already occurring before DLI.

As discussed above, initially DLI was incorpo- rated into NST protocols early in the course for conversion of MC to CC.5MC may be associated with increased risk for relapse, especially in aggressive malignancies, and may also be asso- ciated with MRD. However, as experience with NST was gained, the role of DLI in this setting became more controversial. High-level MC (more than 50%–60% donor chimerism) usually converts to CC spontaneously, whereas patients with low-level donor chimerism (less than 20%–40%) often reject the graft despite DLI. DLI is now administered more carefully for this indi- cation, and patients are followed closely with DLI reserved for impeding graft rejection as evi- denced by declining chimerism. In patients with aggressive malignancies, DLI may still be administered early trying to convert to CC and induce GVL rapidly.

The second indication for DLI has been for persistent or progressive disease, with an overall response rate of 25%–45%, depending on the underlying disease.39,40 DLI has also been explored as prophylactic therapy after NST.

Although this approach may reduce relapse risk, responses are often associated with GVHD.

Because DLI is still associated with substantial risk, a more rational approach is to try to limit DLI only to those destined to relapse, avoiding unnecessary toxicity from those destined to remain in remission based on determination of MRD after NST.42

Significant progress has been achieved in technologies for MRD assessment.42Quantitative

(6)

polymerase chain reaction tests are very sensi- tive in detecting tumor-associated transcripts, allowing serial monitoring. Threshold levels have been established for some malignancies above which relapse is imminent. Persistent negative tests, low level or decreasing MRD level are consistent with continuous remission, whereas, high-level MRD or increasing levels predict incipient relapse. Patients at high risk for relapse are candidates for additional cellular or targeted therapy.

The optimal time point and cell dose of DLI have not been established. The decision to administer DLI can be based on several factors:

The aggressiveness of the underlying malig- nancy and the risk for rapid progression, the sensitivity of the test used to determine MRD, the expected kinetics of MRD and the trend of MRD in serial quantitative testing, the level of MRD, as well as the SCT regimen used.

In indolent malignancies such as chronic myeloid leukemia (CML), chronic lymphocytic leukemia, follicular lymphoma, and to a lesser extent multiple myeloma, MRD is often detected after SCT, both after ablative conditioning, and even more so after reduced-intensity condition- ing. MRD can be followed and no intervention is indicated unless progression or a plateau in response is observed or quantitative MRD is ris- ing. In aggressive malignancies such as acute leukemia and CML in blast crisis, and especially when not in remission at SCT, timing is more crucial. There may not be sufficient time to fol- low quantitative MRD because the doubling time of MRD may be short and relapse may occur within weeks, whereas effective DLI response may take 2–3 months. Thus, the sensi- tivity of the test is important. When using very sensitive tests, such as quantitative polymerase chain reaction, when applicable, one can follow MRD very closely, every 1–2 weeks and if MRD is declining, no intervention is needed. The kinetics of MRD in this setting after NST is not well established as after ablative conditioning.42 The same level of MRD may not necessarily have the same significance. MRD surviving high-dose chemotherapy, and to a lesser extent reduced- intensity conditioning, represent highly resist- ant malignancy, whereas MRD is expected after NST. MRD remaining after T cell-depletion SCT or the use of alemtuzumab in NST is also highly predictive of relapse.

In the future, tumor-specific lymphocytes, or DLI generated against hematopoietic-specific

minor histocompatibility antigens, such as HA- 1 and HA-2,43may be used to harness antitumor responses without the risk of GVHD, and may follow SCT with T cell-depleted grafts.

Targeted therapy is another option for treat- ment or control of MRD. Imatinib mesylate is an effective therapy for CML. There is emerging data that imatinib may be effective in salvaging patients with relapse or persistent disease after SCT, either front line, or as second-line therapy after failure of DLI. Imatinib may also have a synergistic effect with DLI.13 Rituximab is another example. We have used rituximab after SCT in patients with aggressive lymphoma.14 The reduced risk of relapse in very high-risk patients suggested that rituximab may have eliminated MRD. It may have synergized with the donor immune system providing effectors for antibody-dependent cytotoxicity. The MD Anderson group showed similar effects of ritux- imab administered for residual chronic lympho- cytic leukemia after NST.44 Future studies may identify other methods to target MRD, trying to reduce relapse risk after SCT.

Selection of Conditioning Regimen

As a general role, myeloablative conditioning is the standard conditioning before SCT, and NST is still considered an experimental therapy, in which the long-term results have not yet been well defined. As a result, NST should be admin- istered in carefully designed clinical studies.

NST regimens should be reserved mainly for patients not eligible for standard ablative condi- tioning on the basis of the criteria discussed above. However, some groups have also explored NST regimens as a means to reduce toxicity even in younger and medically fit patients in some settings.

The selection of the appropriate regimen for a patient depends on several factors including age, general medical condition, immune compe- tence of the recipient, genetic disparity between the patient and donor, and center experience.2 Perhaps the most important determining factor is the aggressiveness and chemosensitivity of the underlying malignancy and its known suscepti- bility to the GVL effect.

There is now a spectrum of nonmyeloablative and reduced-intensity regimens with different immunosuppressive and cytoreductive intensity.

(7)

The reduced-intensity regimens are a more appropriate approach for aggressive malignan- cies such as acute leukemia, and especially when not in remission. In this setting, rapid achieve- ment of CC and transient disease control is needed to induce GVL. However, in indolent malignancies, GVM may occur slowly, even in mixed chimeras, and toxicity may be reduced further using nonmyeloablative regimens. More intensive immune suppression is required for engraftment of allografts from unrelated donors and in patients not previously treated with chemotherapy. Less intensive immune suppres- sion is needed in heavily pretreated patients and in particular those with a recent prior autolo- gous SCT.2

There is no prospective study comparing the outcomes after different NSTs and reduced- intensity regimens that can show an advantage of one over the others. One analysis showed that reduced-intensity regimens might give better results in disease control than nonmyeloablative conditioning in patients with active or refrac- tory leukemia whereas results were equivalent when the leukemia was in remission at the time of SCT.45 Among reduced-intensity regimens, we have shown that the use of intravenous busulfan is associated with less regimen-related toxicity that other regimens, after both related and unrelated SCT, similar to what we have shown in ablative conditioning.17,46Thus, differ- ences between regimens may be significant and studies comparing them, and then comparing the best regimen to standard ablative regimen, are urgently needed.

CML is the malignancy most sensitive to GVL as evidenced by the high response rates to DLI.

There were some concerns of relatively high inci- dence of graft rejection in patients conditioned with truly nonmyeloablative regimens who had chronic-phase CML and had not been previously treated with intensive chemotherapy.7However, consistent engraftment has been achieved with reduced-intensity conditioning. The Hadassah group reported excellent outcomes in CML in the first chronic phase with 85% of patients surviving disease-free.47Results in advanced-phase CML are much less favorable. We currently recommend reduced-intensity conditioning even for younger patients with chronic-phase CML; however, it should be appreciated that there are no long-term studies prospectively comparing the two approaches. For advanced-phase CML, we would recommend reduced-intensity conditioning only

for those not eligible for ablative conditioning and usually after a trial of remission induction.

Acute leukemia is a more aggressive malig- nancy, which is less susceptible to DLI.

Responses can be achieved; however, they are most often transient. Although there are no ran- domized trials, emerging data suggest that NST may be equivalent or inferior to ablative ther- apy.48,49 However, these comparisons may be biased by the criteria for patient allocation.

Reduced-intensity conditioning may have favor- able results in patients in remission; however, results have been disappointing in patients with active and refractory leukemia. In these patients, leukemia often recurs shortly after NST outpacing the developing of the GVL response.

Acute lymphatic leukemia (ALL) is considered the least responsive disease to immune effects.

This is not because ALL is not susceptible to GVL, rather because ALL is a very rapidly grow- ing malignancy outpacing GVL. Thus, NST may be successful in high-risk ALL in remission, but has a limited role in active disease.

Hodgkin’s and non-Hodgkin’s lymphoma, and multiple myeloma are often treated with autologous SCT. These diseases show moderate susceptibility to GVM effect. When these patients are candidates for allogeneic SCT, they are often heavily pretreated and standard condi- tioning is associated with unacceptably high TRM rates. NST is becoming the preferred approach in these patients. It is feasible with favorable results in chemosensitive diseases.50 Multiple myeloma is an incurable disease with standard chemotherapy as well as with autolo- gous SCT. NST is being explored as an approach that may achieve cure. It is now established that relapse rates are very high when NST is given after relapse to prior therapy, but may be lower when NST is given upfront. The auto/allo approach in which autologous SCT is given for cytoreduction and is followed by NST to induce GVM seems promising in this setting.12

Chronic lymphatic leukemia and follicular lymphoma have been shown to be very sensitive to immune effects, similar to CML. These are indolent diseases for which standard ablative conditioning have very high TRM rates. Thus, these diseases are reasonable targets for NST, and we currently prefer this approach, especially in patients with chemosensitive disease.8,44 As discussed above, SCT can also be supplemented with targeted therapy, such as rituximab, both before and after NST in these diseases.

(8)

Conclusions

Nonmyeloablative and reduced-intensity condi- tioning are increasingly being used before allo- geneic SCT, in a growing number of indications.

It is now well established that the first goal of allowing SCT for elderly and medically infirm patients has been achieved. NST regimens result in consistent engraftment of allografts from related and unrelated donors. TRM rates have been markedly reduced such that SCT can be administered relatively safely with no upper age limit, and after prior autologous SCT, as well as in certain malignancies, such as lymphomas and myeloma where historically TRM rates were exceedingly high. However, toxicity may still be substantial with some regimens and in patients with a high comorbidity score. GVHD continues to be a major cause of morbidity and mortality after NST, and its incidence may not be lower than after ablative SCT. Invasive fungal infec- tions are a second common cause of TRM, which is closely associated with GVHD, and did not reduce in incidence. Novel approaches to further reduce these two complications are required to further improve outcome. In vivo T cell depletion reduces initial rates of GVHD, but because DLI is required more often for increased risk of disease persistence and MC, the ultimate rate of GVHD remains unchanged.

With gained experience, DLI is used more care- fully after NST, limiting its use to patients with persistent MRD after SCT, or imminent graft rejection, and delaying administration in others, trying to reduce the risk for GVHD. Methods to deliver cellular immune therapy without GVHD would be a major step forward. The develop- ment of tumor or minor histocompatibility anti- gen restricted DLI, and the combination with targeted therapy and tumor vaccines are prom- ising. Currently, despite initial data, there is no firm evidence for advantage of any of the regi- mens over the others. Although TRM may reduce with NST, theoretically, relapse rates may increase compared with ablative SCT, such that the net effects on disease-free survival are yet to be determined. Prospective comparative studies to determine the best NST regimen and then randomized studies comparing NST and ablative SCT, are urgently required before NST can be accepted as standard therapy, and to better define its role.

References

1. Thomas ED, Storb R, Clift RA, et al. Bone marrow transplantation. N Engl J Med 1975;292:832–843.

2. Champlin R, Khouri, Shimoni A, et al. Harnessing graft- versus-malignancy: nonmyeloablative preparative regi- mens for allogeneic haematopoietic transplantation.

An evolving strategy for adoptive immunotherapy.

Br J Haematol 2000;11:18–29.

3. Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alterna- tive to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and non malignant hematologic diseases. Blood 1998;91:

756–763.

4. Giralt S, Estey E, Albitar M, et al. Engraftment of allo- geneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft- versus-leukemia without myeloablative therapy.

Blood 1997;89:4531–4536.

5. Shimoni A, Nagler A. Non-myeloablative stem cell transplantation (NST): chimerism testing as guidance for immune-therapeutic manipulations. Leukemia 2001;15:1967–1975.

6. Bacigalupo A, Van Lint MT, Occhini D, et al. Increased risk of leukemia relapse with high-dose cyclosporine A after allogeneic transplantation for acute leukemia.

Blood 1991;77:1423–1428.

7. McSweeney PA, Niederwieser D, Shizuru JA, et al.

Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects.

Blood 2001;97:3390–3400.

8. Khouri I, Keating MJ, Korbling M, et al. Transplant lite:

induction of graft vs malignancy using fludarabine based nonablative chemotherapy and allogeneic pro- genitor-cell transplantation as treatment for lymphoid malignancies. J Clin Oncol 1998;16:2817–2824.

9. Giralt S, Thall PF, Khouri I, et al. Melphalan and purine analog-containing preparative regimens: reduced- intensity conditioning for patients with hematologic malignancies undergoing allogeneic progenitor cell transplantation. Blood 2001;97:631–637.

10. Perez-Simon JA, Kottaridis PD, Martino R, et al.

Nonmyeloablative transplantation with or without alemtuzumab: comparison between 2 prospective studies in patients with lymphoproliferative disorders.

Blood 2002;100:3121–3127.

11. Carella AM, Cavaliere M, Lerma E, et al. Autografting followed by nonmyeloablative immunosuppressive chemotherapy and allogeneic peripheral blood hematopoietic stem-cell transplantation as treatment of resistant Hodgkin’s disease and non-Hodgkin’s lym- phoma. J Clin Oncol 2000;18:3918–3924.

12. Kroger N, Schwerdtfeger R, Kiehl M, et al. Autologous stem cell transplantation followed by a dose-reduced allograft induces high complete remission rate in mul- tiple myeloma. Blood 2002;100:755–760.

13. Shimoni A, Kroger N, Zander AR, et al. Imatinib mesy- late (STI571) in preparation for allogeneic hematopoi- etic stem cell transplantation and donor lymphocyte infusions in patients with Philadelphia-positive acute leukemias. Leukemia 2003;17:290–297.

(9)

14. Shimoni A, Hardan I, Avigdor A, et al. Rituximab reduces relapse risk after allogeneic and autologous stem cell transplantation in patients with high-risk aggressive non-Hodgkin’s lymphoma. Br J Haematol 2003;122:457–464.

15. Sandmaier BM, Bethge WA, Wilbur DS, et al. Bismuth 213-labeled anti-CD45 radioimmunoconjugate to con- dition dogs for nonmyeloablative allogeneic marrow grafts. Blood 2002;100:318–326.

16. Corradini P, Zallio F, Mariotti J, et al. Effect of age and previous autologous transplantation on treatment- related mortality and graft-versus-host disease in 110 patients treated with reduced-intensity conditioning and allografting for advanced hematological malig- nancies [abstract]. Bone Marrow Transplant 2004;33 (suppl 1):S11.

17. Shimoni A, Kroger N, Zander A, et al. Allogeneic stem- cell transplantation with matched unrelated donors in he elderly (age >55). Age is no longer a contraindica- tion when using reduced-intensity conditioning.

Leukemia 2004. In press.

18. Nagler A, Or R, Naparstek E, Varadi G, Slavin S. Second allogeneic stem cell transplantation using non-mye- loablative conditioning for patients who relapsed or developed secondary malignancies following autolo- gous transplantation. Exp Hematol 2000;28:1096–1104.

19. Feinstein LC, Sandmaier BM, Maloney DG, et al.

Allografting after nonmyeloablative conditioning as a treatment after a failed conventional hematopoietic cell transplant. Biol Blood Marrow Transplant 2003;9:

266–272.

20. Nagler A, Aker M, Or R, et al. Low-intensity condition- ing is sufficient to ensure engraftment in matched unrelated bone marrow transplantation. Exp Hematol 2001;29:1–9.

21. Maris MB, Niederwieser D, Sandmaier BM, et al. HLA- matched unrelated donor hematopoietic cell transplan- tation after nonmyeloablative conditioning for patients with hematologic malignancies. Blood 2003;102:

2021–2030.

22. Diaconescu R, Flowers C, Storer B, et al. Morbidity and mortality with nonmyeloablative compared to mye- loablative conditioning before hematopoietic cell transplantation from HLA matched related donors.

Blood 2004;104:1550–1558.

23. Fukuda T, Hackman RC, Guthrie KA, et al. Risks and outcomes of idiopathic pneumonia syndrome after nonmyeloablative and conventional conditioning regi- mens for allogeneic hematopoietic stem cell transplan- tation. Blood 2003;102:2777–2785.

24. Hogan WJ, Maris M, Storer B, et al. Hepatic injury after nonmyeloablative conditioning followed by allogeneic hematopoietic cell transplantation: a study of 193 patients. Blood 2004;103:78–84.

25. Shimoni A, Yeshurun M, Hardan H, et al. Thrombotic microangiopathy following allogeneic stem-cell trans- plantation in the era of reduced-intensity conditioning:

the incidence is not reduced. Biol Bone Marrow Transplant 2004;10:484–493.

26. Worel N, Kalhs P, Keil F, et al. ABO mismatch increases transplant-related morbidity and mortality in patients given nonmyeloablative allogeneic HPC transplanta- tion. Transfusion 2003;43:1153–1161.

27. Sorror ML, Maris M, Storer B, et al. Transplant-related toxicities (TRT) and mortality following HLA-matched unrelated donor hematopoietic cell transplantation

(URD-HCT) using nonmyeloablative (NM) compared to myeloablative (M) conditioning: influence of pre- transplant comorbidities. Blood 2004;104:961–968.

28. Weissinger F, Sandmaier BM, Maloney DG, et al.

Decreased transfusion requirements for patients receiving nonmyeloablative compared with conven- tional peripheral blood stem cell transplants from HLA-identical siblings. Blood 2001;98:3584–3588.

29. Busca A, Locatelli F, Barbui A, et al. Infectious com- plications following nonmyeloablative allogeneic hematopoietic stem cell transplantation. Transpl Infect Dis 2003;5:132–139.

30. Junghanss C, Marr KA, Carter RA, et al. Incidence and outcome of bacterial and fungal infections following nonmyeloablative compared with myeloablative allo- geneic hematopoietic stem cell transplantation: a matched control study. Biol Blood Marrow Transplant 2002;8:512–520.

31. Fukuda T, Boeckh M, Carter RA, et al. Risks and out- comes of invasive fungal infections in recipients of allo- geneic hematopoietic stem cell transplants after nonmyeloablative conditioning. Blood 2003;102:827–833.

32. Hagen EA, Stern H, Porter D, et al. High rate of invasive fungal infections following nonmyeloablative allo- geneic transplantation. Clin Infect Dis 2003;36:9–15.

33. Antin JH, Ferrara JLM. Cytokine dysregulation and acute graft-vs-host disease. Blood 1992;80:2964–2968.

34. Prigozhina T, Gurevitch O, Slavin S. Non-myeloabla- tive conditioning to induce bilateral tolerance after allogeneic bone marrow transplantation in mice. Exp Hematol 1999;27:1503–1510.

35. Shlomchik WD, Couzens MS, Tang CB, et al. Prevention of graft versus host disease by inactivation of host anti- gen-presenting cells. Science 1999;285:412–415.

36. Naparstek E, Or R, Nagler A, et al. T-cell depleted allo- geneic bone marrow transplantation for acute leukemia using Campath-1 antibodies and post-transplant admin- istration of donor’s peripheral blood lymphocytes for prevention of relapse. Br J Haematol 1995;89:506–515.

37. Mielcarek M, Martin PJ, Leisenring W, et al. Graft-ver- sus-host disease after nonmyeloablative versus con- ventional hematopoietic stem cell transplantation.

Blood 2003;102:756–762.

38. Couriel DR, Saliba RM, Giralt S, et al. Acute and chronic graft-versus-host disease after ablative and nonmye- loablative conditioning for allogeneic hematopoietic transplantation. Biol Blood Marrow Transplant 2004;10:178–185.

39. Marks DI, Lush R, Cavenagh J, et al. The toxicity and efficacy of donor lymphocyte infusions given after reduced-intensity conditioning allogeneic stem cell transplantation. Blood 2002;100:3108–3114.

40. Bethge WA, Hegenbart U, Stuart MJ, et al. Adoptive immunotherapy with donor lymphocyte infusions after allogeneic hematopoietic cell transplantation following nonmyeloablative conditioning. Blood 2004;

103:790–795.

41. Mapara MY, Kim YM, Wang SP, et al. Donor lympho- cyte infusions mediate superior graft-versus-leukemia effects in mixed compared to fully allogeneic chimeras:

a critical role for host antigen-presenting cells. Blood 2002;100:1903–1909.

42. Shimoni A, Nagler A. Clinical implications of MRD monitoring for stem-cell transplantation after reduced-intensity and non-myeloablative condition- ing. Acta Haematol 2004;112:93–104.

(10)

43. Marijt WA, Heemskerk MH, Kloosterboer FM, et al.

Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia. Proc Natl Acad Sci USA 2003;100:2742–2747.

44. Khouri IF, Lee MS, Saliba RM, et al. Nonablative allo- geneic stem cell transplantation for chronic lymphocytic leukemia: impact of rituximab on immunomodulation and survival. Exp Hematol 2004;32:28–35.

45. Shimoni S, Giralt S, Khouri I, Champlin R. Allogeneic hematopoietic transplantation for acute and chronic myeloid leukemia: non-myeloablative preparative reg- imens and induction of the graft-versus-leukemia effect. Curr Oncol Rep 2000;2:132–139.

46. Shimoni A, Hardan I, Yeshurun M, et al. Intravenous busulfan versus melphalan-based reduced intensity conditioning prior to allogeneic stem cell transplan- tation: lower TRM and a more favorable toxicity pro- file [abstract]. Bone Marrow Transplant 2004;33 (suppl 1):S12.

47. Or R, Shapira MY, Resnick I, et al. Nonmyeloablative allogeneic stem cell transplantation for the treatment

of chronic myeloid leukemia in first chronic phase.

Blood 2003;101:441–445.

48. Alyea EP, Kim HT, Cutler C, et al. AML and MDS treated with nonmyeloablative stem cell transplanta- tion: overall and progression free survival comparable to myeloablative transplantation [abstract]. Blood 2003;102:79a.

49. Fouillard L, Labopin M, Rocha V, et al. Comparison of reduced-intensity conditioning regimen to conven- tional conditioning regimen in allogeneic haematopoi- etic stem cell transplantation for acute myeloid leukemia [abstract]. Bone Marrow Transplant 2004;33 (suppl 1):S13.

50. Robinson SP, Goldstone AH, Mackinnon S, et al.

Chemoresistant or aggressive lymphoma predicts for a poor outcome following reduced-intensity allogeneic progenitor cell transplantation: an analysis from the Lymphoma Working Party of the European Group for Blood and Bone Marrow Transplantation. Blood 2002;100:4310–4316.

Riferimenti

Documenti correlati

For the class of nonlinear control-affine systems, both differential equations and inclusions, we have shown the equivalence between the control-sharing property and the existence

The exceptionally high intensity of the ultraplinian regime, and the very large amount of fine ash generated during magma fragmentation, results in convective columns up to 55 km

(2015) Investigating the Role of the Host Multidrug Resistance Associated Protein Transporter Family in Burkholderia cepacia Complex Pathogenicity Using a Caenorhabditis

To the best of our knowledge, there are no studies assessing the effectiveness of a specific training for the communicative- pragmatic abilities in patients with schizophrenia, using

The archaeometric study performed on the ‘The divine adoration of King St. Louis IX’ fresco identified two causes as the main responsible for the darkening and loss of

Tanto per prospettiva che per utilizzo delle fonti, il contributo ripropone dunque il punto di vista privilegiato dello Stato etiopico come un “centro” che gradualmente ingloba