Hematopoietic Cell Transplantation


Hematopoietic cell transplantation is a procedure by which hematopoietic stem and progenitor cells are infused intravenously to restore hematopoiesis and immune function following a chemotherapy conditioning regimen, with or without radiation therapy, that ablates the endogenous hematopoietic and immune system. Stem cells used for hematopoietic cell transplantation are of hematopoietic origin, in contrast to the more primitive pluripotent stem cells (i.e., embryonic stem cells) that are considered for regenerative therapy because of their ability to differentiate into virtually any somatic cell. Hematopoietic cell transplantation is now a standard treatment for many hematologic malignancies, immunodeficiency states, enzyme deficiencies (e.g., Hurler syndrome [ Chapter 192 ]), and defective hematopoietic states (e.g., severe aplastic anemia [ Chapter 151 ]). In addition, hematopoietic cell transplantation can also treat hemoglobinopathies, immune deficiency states, and selected autoimmune diseases. The infused hematopoietic stem/progenitor cells can give rise to all blood elements, including cells of the immune system, and they may be genetically modified to enhance their function. Furthermore, hematopoietic stem/progenitor cells may be manipulated ex vivo and used in the burgeoning field of regenerative medicine. Taken together, hematopoietic cell transplantation is a major component and foundation of the broader field of cellular therapy ( Chapter 34 ).

The three cell sources for hematopoietic cell transplantation are syngeneic (i.e., cells from an identical twin), allogeneic (i.e., cells from a nonidentical donor), and autologous (i.e., reinfusion of one’s own cells). The determination of the type of hematopoietic cell transplantation that a patient receives is based on the disease to be treated, the disease state (e.g., initial treatment vs. treatment of recurrent disease), the urgency with which the disease needs to be treated, the availability of a donor, and the time necessary to obtain hematopoietic stem/progenitor cells from a donor.

Hematopoietic stem/progenitor cells may be obtained from bone marrow (by serial aspirations from the posterior iliac crests while the donor is under general anesthesia), peripheral blood (after stimulation with hematopoietic growth factors such as granulocyte colony–stimulating factor [G-CSF] or plerixafor [a CXCR4 antagonist], followed by leukapheresis), or umbilical cord blood. In current practice, peripheral blood is the source for hematopoietic stem/progenitor cells in approximately 90% of autologous transplants and in approximately 70% of allogeneic transplants because of the relative ease of procurement and the donor’s rate of hematopoietic recovery after infusion compared with cells derived from bone marrow.

Hematopoietic stem/progenitor cells may be infused fresh or after cryopreservation, which involves processing the cells in culture medium containing dimethyl sulfoxide (DMSO), placing them in specialized plastic bags, controlled freezing, and then storing in the vapor phase of liquid nitrogen until needed. Cord blood hematopoietic stem/progenitor cells are collected at the time of delivery, cryopreserved, and stored. For allogeneic donation, cells may also be infused on the day of collection. For autologous donation, cells are almost always cryopreserved.

Allogeneic Hematopoietic Cell Transplantation

The distinctive characteristics of allogeneic hematopoietic cell transplantation are that the stem cell graft allows for the establishment of a new immune system in addition to reconstitution of white blood cells, red blood cells, and platelets. The new immune system can be a major advantage if the immunologic response is directed against malignant cells, referred to as the graft-versus-leukemia or graft-versus-tumor effect, which potentially can eradicate disease and reduce the chance of disease relapse. However, if the immunologic response is directed against antigens present on normal tissues, the graft-versus-host response can result in the destruction of normal organs and is clinically described as graft-versus-host disease (GVHD). The risk for both graft rejection (host-versus-graft reaction) and GVHD rises with the degree of human leukocyte antigen (HLA) disparity between the donor and the recipient as well as the degree of immune suppression.

The clinical importance of the role that immunocompetent donor T cells play in mediating a graft-versus-tumor effect is exemplified by the increased rates of relapse in patients who receive allogeneic bone marrow grafts from which T cells had been removed (T-cell depletion), an inverse correlation between relapse and severity of GVHD, and increased rates of relapse after syngeneic or autologous hematopoietic cell transplantation in patients who have received similar conditioning regimens. However, the most compelling evidence for the importance of T cells mediating the graft-versus-tumor effect is the clinical observation that infusion of allogeneic T cells alone, termed a donor lymphocyte infusion , at a time remote from the transplant conditioning regimen can successfully eradicate leukemia that has persisted or recurred after allogeneic hematopoietic cell transplantation. Nevertheless, the clinical effectiveness of graft-versus-tumor can vary widely among different malignancies.

Because of the immunologic barriers that can result in graft rejection and GVHD, allogeneic hematopoietic cell transplantation requires that the donor and the recipient share as many key genes as possible. The most important of these are HLAs ( Chapter 38 ), which are derived from the MHC located on chromosome 6. The HLA loci include HLA-A, HLA-B, HLA-C, DR, DP, and DQ. A single set of major histocompatibility complex (MHC) alleles, described as a haplotype, is inherited from each parent, thereby resulting in HLA pairs. A clinical “match” means that HLAs in the donor match the HLAs in the patient.

The choice of donor for an allogeneic hematopoietic cell transplantation takes into account several factors, including the patient’s specific disease and disease state (i.e., remission vs. relapse) and the time that is required for obtaining hematopoietic stem/progenitor cells from a donor. A fully HLA-matched sibling is the preferred donor source because the risks for graft rejection and GVHD are lowest. The probability of having an HLA match with a single sibling is 25% and increases with the number of siblings within a specific family. This probability can be estimated using the following formula: chance of having an HLA-matched sibling = 1 − (0.75) n , where n is the number of potential sibling donors. There is approximately a 1% chance of a crossover event (i.e., genetic material switched between chromosomes during meiosis), primarily between the HLA-A and the HLA-B loci. The clinical outcomes for allogeneic hematopoietic cell transplantation using a sibling with a single HLA mismatch are similar to those with a fully HLA-matched sibling.

For patients who lack an HLA-identical sibling donor, the alternative sources for allogeneic hematopoietic stem/progenitor cells include a fully HLA-matched volunteer unrelated donor, a fully or partially HLA-matched cord blood unit, or a partially HLA-matched first-degree family member. HLA genes are highly polymorphic, so the odds that any two unrelated individuals are HLA identical for main loci are less than 1 in 10,000. However, tens of millions of people worldwide are currently listed as potential volunteer donors, so a donor can be found for about 50% of all patients for whom a search is initiated. The probability of identifying a suitable volunteer donor is highly dependent on race because of varying degrees of HLA diversity. For example, for persons of northern European extraction, the probability of a match can be as high as 90%. In contrast, minority populations may have less than a 20% probability of a match because of fewer volunteer donors and a higher degree of genetic variation.

It usually takes about 2 to 4 months to locate and fully evaluate an unrelated donor, which may be too long for some patients with rapidly progressive malignancies. When a suitable volunteer is not available or a donor is needed more urgently, alternative sources can be considered, including partially HLA-matched (“haploidentical”) relatives or umbilical cord blood. Haploidentical hematopoietic stem/progenitor cells are readily available but have increased risks for graft rejection and GVHD. These risks can be reduced by either ex vivo or in vivo T-cell depletion methods, but T-cell depletion may lead to delayed immune reconstitution and competency after transplantation, thereby increasing the risks for infection and recurrent disease. However, post-transplantation cyclophosphamide for GVHD prophylaxis has markedly increased the clinical use of haploidentical donors.

Umbilical cord blood hematopoietic stem/progenitor cells, which are taken at the time of delivery and stored in cord blood banks, are readily available within 2 to 4 weeks when needed. Because of the unique immature biology of lymphocytes in umbilical cord blood, these transplants are associated with less GVHD; consequently, the HLA-matching requirements are less strict, and two or three HLA mismatches may be acceptable. However, the small volume (50 to 150 mL) of cord blood that can be collected limits the number of hematopoietic stem/progenitor cells and often prohibits their use in adults because successful engraftment correlates with the number of cells per patient body weight. The limitation of cell dose can be overcome by the use of more than one cord blood unit and the ability to expand cord blood cells ex vivo, thereby resulting in engraftment rates similar to other cell sources. One significant disadvantage is that the use of a cord blood unit eliminates the probability of obtaining additional cells if the graft fails or if a donor lymphocyte infusion is required.

After a source of allogeneic stem cells has been identified, patients then receive a pretransplant regimen with the intent of “conditioning” or “preparing” them for the infusion ( Fig. 163-1 ). These conditioning regimens are designed to be adequately immunosuppressive to overcome the host-versus-graft reaction and permit engraftment. They also are designed to eradicate the primary tumor in patients with an underlying malignancy. Most conditioning regimens use chemotherapy alone or a combination of total body irradiation or total nodal irradiation with chemotherapy. The doses of total body and total lymphoid irradiation vary between 200 and 1440 cGy. The most commonly used chemotherapy for conditioning regimens is an alkylating agent (e.g., cyclophosphamide, busulfan, thiotepa) combined with immunosuppressive drugs such as fludarabine. Conditioning regimens also may contain monoclonal antibodies that target T cells (e.g., antithymocyte globulin, alemtuzumab).

FIGURE 163-1, The process of allogeneic hematopoietic cell transplantation.

The choice of a specific conditioning regimen depends on the disease being treated, and the doses of chemotherapy and radiation are highly variable. When the doses result in a degree of myelosuppression and immunosuppression that is nearly universally fatal without the infusion of hematopoietic stem/progenitor cells as a rescue product, they are referred to as myeloablative.

Allogeneic hematopoietic cell transplantation with myeloablative conditioning regimens has been performed successfully in patients older than age 60 years, but survival after transplantations declines with increasing age, thereby limiting the application of allogeneic transplantation to a minority of patients who potentially could benefit from this procedure. However, the demonstration that an immune-mediated graft-versus-tumor effect plays a central role in the therapeutic efficacy of allogeneic hematopoietic cell transplantation led to the development of less intensive conditioning regimens that are adequately immunosuppressive to permit the engraftment of donor cells but associated with decreased toxicities compared with myeloablative regimens. Another advantage of less intensive conditioning is that it can be delivered outside of the hospital setting, so that patients receive their care in the outpatient setting and are able to stay in a local apartment or their actual home if they live close to the center. Although the reduced doses of radiation and chemotherapy result in decreased antitumor activity and are associated with higher rates of recurrent disease after transplantation, these “nonmyeloablative” and “reduced-intensity” conditioning regimens have markedly increased the potential option of allogeneic hematopoietic cell transplantation for older patients and patients with preexisting comorbidities.

Syngeneic Hematopoietic Cell Transplantation

The major advantage of a syngeneic hematopoietic cell transplantation from an identical twin is that it is not associated with GVHD or graft rejection, thereby resulting in a relatively low risk for treatment-related morbidity and mortality. Syngeneic hematopoietic cell transplantation, as with allogeneic transplantation, also avoids potential contamination with malignant cells. The major disadvantage of syngeneic transplantation is that it does not provide the graft-versus-leukemia effect associated with allogeneic transplantation. However, since far less than 1% of patients have an identical twin, syngeneic transplantation is rarely an option.

Autologous Hematopoietic Cell Transplantation

The major rationale for autologous hematopoietic cell transplantation with the patient’s own cells is that certain malignancies, such as leukemias and lymphomas, have a steep dose-response curve to chemotherapy and, to a relative degree, radiation. However, the associated limitation of higher doses of chemotherapy or radiation attenuates myelosuppressive effect. The infusion of autologous hematopoietic stem/progenitor cells (and for that matter, allogeneic and syngeneic cells) can restore hematopoiesis after the prior administration of high-dose chemotherapy, with or without radiation. The major advantages of autologous as compared with allogeneic transplantation are that the patient can serve as his or her own donor and that it may be performed in older patients with significantly lower morbidity and mortality because of the absence of GVHD. Although patients undergoing autologous hematopoietic cell transplantation have higher relapse rates than patients undergoing allogeneic transplantation, the lower rates of other complications seem to translate into similar long-term outcomes. Nevertheless, the higher conditioning doses of chemotherapy before autologous hematopoietic cell transplantation can be associated with more morbidity than conventional doses of chemotherapy.

Indications for Transplantation

An estimated 90,000 hematopoietic cell transplantations are performed annually worldwide, primarily for the treatment of hematologic malignancies, bone marrow failure states, and immune and enzyme deficiencies ( Fig. 163-2 ).

FIGURE 163-2, Indications for hematopoietic cell transplantation in the United States as reported to the Center for International Blood and Marrow Transplant Research.

Acute Myeloid Leukemia

Allogeneic hematopoietic cell transplantation is capable of resulting in the long-term survival of 10% of patients with otherwise refractory acute myeloid leukemia (AML; Chapter 168 ). By comparison, cure rates of 40 to 70% have been reported with hematopoietic cell transplantation performed during a first complete remission, and rates of 20 to 40% have been achieved in patients treated while in a second or subsequent complete remission. In randomized trials of patients with AML in first complete remission, both autologous and allogeneic hematopoietic cell transplantation provide improved leukemia-free survival compared with conventional chemotherapy. In terms of overall survival, however, meta-analyses demonstrate improvement with allogeneic hematopoietic cell transplantation compared with nonallogeneic treatments for AML in first complete remission in patients who have intermediate- and high-risk cytogenetics but not in patients who have good-risk AML. Data also indicate the importance of molecular mutations. For example, intermediate-risk AML with a normal karyotype represents a highly heterogeneous group with respect to prognosis based on molecular mutation status. Approximately one third of patients with normal-karyotype AML harbor the FLT3 -internal tandem mutation, which carries a poor prognosis and may benefit from HLA-matched related hematopoietic cell transplantation, regardless of the presence of other mutations.

Acute Lymphoblastic Leukemia

Conventional chemotherapy provides excellent outcomes for childhood acute lymphoblastic leukemia (ALL), except for ALL associated with the Philadelphia chromosome ( Chapter 168 ). In adults, however, remission, though frequently attained after intensive induction therapy, is commonly followed by relapse. Adverse prognostic factors include the presence of the Philadelphia chromosome, a high white blood cell count, advancing age, and the presence of minimal residual disease. Studies suggest that consolidation therapy including hematopoietic cell transplantation after initial chemotherapy improves overall survival compared with conventional chemotherapy alone, particularly for those who are Philadelphia chromosome–positive. For both pediatric and adult patients with relapsed ALL, the overall prognosis is relatively poor, and the general treatment strategy is to obtain a second complete remission and then proceed to an allogeneic hematopoietic cell transplantation.

Myelodysplastic Syndrome

The only known curative treatment for myelodysplastic syndrome ( Chapter 167 ) is allogeneic hematopoietic cell transplantation. Unfortunately, the vast majority of patients are not considered candidates for this therapy for many reasons, including advanced age (the median age at diagnosis is in the seventh to eighth decade of life), comorbid diseases, the inability to identify a suitable donor, and the patient’s preferences. The best results have been obtained in younger patients, who are earlier in their disease course and have not received any prior therapy. Increasing evidence suggests that reduced-intensity allogeneic hematopoietic cell transplantation may benefit even considerably older patients with myelodysplastic syndrome. A randomized trial of patients with myelodysplastic syndrome or secondary AML showed that a busulfan-based reduced-intensity conditioning regimen resulted in at least a 2-year relapse-free and overall survival, similar to that after a myeloablative conditioning regimen for allogeneic stem cell transplantation.

Chronic Myeloid Leukemia

In patients with chronic myeloid leukemia (CML; Chapter 170 ), hematopoietic cell transplantation is reserved for patients who are resistant to or intolerant of treatment with tyrosine kinase inhibitors, especially those who develop an accelerated phase or blast crisis CML. Although results of allogeneic hematopoietic cell transplantation are poor for accelerated-phase or blast-crisis disease, some patients may respond to tyrosine kinase inhibitors after transplantation.

Myeloproliferative Neoplasms

Myeloproliferative neoplasms, such as primary myelofibrosis, polycythemia vera, and essential thrombocythemia ( Chapter 152 ), usually are chronic in nature but can progress to a “spent” phase and evolve into myeloid metaplasia, which is characterized by bone marrow fibrosis and a generally poor prognosis with transformation into acute leukemia and a median survival of less than 3 years. Conventional treatment options are limited at this stage, and the accepted standard of care for myeloid metaplasia/myelofibrosis is allogeneic hematopoietic cell transplantation, especially nonmyeloablative transplantation because of the older age of most patients with this condition.

Non-Hodgkin Lymphoma

Syngeneic, allogeneic, and autologous hematopoietic cell transplantation have all been reported to yield long-term, disease-free survival and an apparent cure for patients with advanced non-Hodgkin lymphoma ( Chapter 171 ). Based on its risk-benefit ratio and availability, the standard of care for patients with primary refractory or chemotherapy-sensitive relapsed non-Hodgkin lymphoma of specific histologies (including diffuse large B-cell lymphoma) has been autologous hematopoietic cell transplantation. For mantle cell lymphoma, autologous hematopoietic cell transplantation appears to improve progression-free and possibly overall survival when used as part of front-line therapy, especially when combined with maintenance rituximab post-transplantation. The development of chimeric antigen receptor (CAR) T cells for lymphomas may challenge this sequence depending on the results of ongoing trials.

However, the graft-versus-tumor effect against various non-Hodgkin lymphoma histologies is very heterogeneous, and consensus is currently lacking regarding for which histology allogeneic hematopoietic cell transplantation is appropriate. For example, autologous transplantation can result in disease-free survival rates as high as 60% in patients with indolent follicular non-Hodgkin lymphoma, but late relapses after transplantation and the long overall survival observed with current medical therapy have reduced the role for hematopoietic cell transplantation despite its graft-versus-tumor effects.

Hodgkin Lymphoma

Based on a small prospective randomized trial conducted in the early 1990s, autologous hematopoietic cell transplantation has become the standard of care for patients with primary refractory and relapsed Hodgkin lymphoma ( Chapter 172 ) whose disease nevertheless remains sensitive to chemotherapy. The usual approach is first to treat these patients with second-line chemotherapy and then with high-dose chemotherapy and autologous hematopoietic cell transplantation. Allogeneic transplantation has had a limited role because of the efficacy of autologous transplantation and the significant treatment-related toxicities associated with myeloablative allogeneic transplantation, but reduced-intensity allogeneic transplantation can be an option, especially in patients who relapse after autologous transplantation.

Multiple Myeloma

Although immunomodulatory derivatives and proteasome inhibitors are highly effective for the treatment of multiple myeloma ( Chapter 173 ), early autologous hematopoietic cell transplantation, when feasible, remains the standard of care. Prospective comparisons of single versus tandem autologous transplants give conflicting results, although overall survival rates appear similar. Single-arm and randomized trials support the use of a second autologous transplantation for patients who relapse after the first autologous transplantation, provided the duration of response was more than 1 year after the initial transplantation.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here